Abstract
The in vitro metabolism of tolperisone, 1-(4-methyl-phenyl)-2-methyl-3-(1-piperidino)-1-propanone-hydrochloride, a centrally acting muscle relaxant, was examined in human liver microsomes (HLM) and recombinant enzymes. Liquid chromatography-mass spectrometry measurements revealed methyl-hydroxylation (metabolite at m/z 261; M1) as the main metabolic route in HLM, however, metabolites of two mass units greater than the parent compound and the hydroxy-metabolite were also detected (m/z 247 and m/z 263, respectively). The latter was identified as carbonyl-reduced M1, the former was assumed to be the carbonyl-reduced parent compound. Isoform-specific cytochrome P450 (P450) inhibitors, inhibitory antibodies, and experiments with recombinant P450s pointed to CYP2D6 as the prominent enzyme in tolperisone metabolism. CYP2C19, CYP2B6, and CYP1A2 are also involved to a smaller extent. Hydroxymethyl-tolperisone formation was mediated by CYP2D6, CYP2C19, CYP1A2, but not by CYP2B6. Tolperisone competitively inhibited dextromethorphan O-demethylation and bufuralol hydroxylation (Ki = 17 and 30 μM, respectively). Tolperisone inhibited methylp-tolyl sulfide oxidation (Ki = 1200 μM) in recombinant flavin-containing monooxygenase 3 (FMO3) and resulted in a 3-fold (p < 0.01) higher turnover number using rFMO3 than that of control microsomes. Experiments using nonspecific P450 inhibitors—SKF-525A, 1-aminobenzotriazole, 1-benzylimidazole, and anti-NADPH-P450-reductase antibodies—resulted in 61, 47, 49, and 43% inhibition of intrinsic clearance in HLM, respectively, whereas hydroxymethyl-metabolite formation was inhibited completely by nonspecific chemical inhibitors and by 80% with antibodies. Therefore, it was concluded that tolperisone undergoes P450-dependent and P450-independent microsomal biotransformations to the same extent. On the basis of metabolites formed and indirect evidences of inhibition studies, a considerable involvement of a microsomal reductase is assumed.
Tolperisone, a centrally acting muscle relaxant (Pórszász et al., 1961), has been used for the treatment of painful muscle spasms in orthopaedic and rheumatologic diseases in several decades. Ono and colleagues have shown that tolperisone exerts a local anesthetic-like action both on motoneurons and primary afferents in vivo, as well as on peripheral nerves of rats in vitro (Ono et al., 1984). The effect of tolperisone is similar to that of lidocaine, which is known to inhibit voltage-dependent sodium currents (Oortgiesen et al., 1990). In a recent study it was shown that tolperisone was effective in patients suffering from painful reflex muscle spasms (Pratzel et al., 1996).
Although a few papers are available on the pharmacokinetics of tolperisone, no study has been performed to date on the in vitro metabolism of the compound. In early in vivo studies the drug concentration in the plasma was measured using gas chromatography and by mass fragmentographic techniques (Miyazaki et al., 1972, 1975). Miskolczi and coworkers in 1987 published a new and sensitive gas-liquid chromatographic method for the determination of tolperisone in human plasma and made a comparison of two different brands of tolperisone tablets, Mydeton and Mydocalm. In that study the elimination half-life of tolperisone was 1.55 ± 0.7 h, and the total body clearance was 140.8 ± 33.8 l/h. The oral bioavailability of the two different tolperisone products was 22.3 ± 6% and 16.7 ± 8.9%, respectively.
Metabolic profiling is a useful tool to predict the pharmacokinetic properties of drugs and drug candidates and to assess the possibility of drug-drug interaction risks. The genetic background and actual metabolic state of individuals can vary largely even in the same population group. Therefore, these differences can influence significantly the fate of ingested drugs (i.e., the AUC1 can differ largely in different individuals). On one hand, the elevated plasma level of a drug can cause toxicity problems, especially those with narrow therapeutic range. On the other hand, high metabolic activity in certain population groups (extensive metabolizers) may prevent therapeutic efficacy of drugs. The goal of the present study was to identify by in vitro reaction phenotyping methods the microsomal enzymes that contribute to the biotransformation of tolperisone and to elucidate the metabolic profile of the compound.
Materials and Methods
Chemicals.
Tolperisone-hydrochloride and metabolite standards were synthesized in Gedeon Richter Ltd. (Budapest, Hungary). Quinidine was purchased from Merck (Darmstadt, Germany); ketoconazole, sulfaphenazole, 6β-hydroxytestosterone, SKF-525A (proadifen-hydrochloride), glucose 6-phosphate, glucose-6-phosphate dehydrogenase, phenacetin, 4-acetamidophenol and 1-aminobenzotriazole were purchased from Sigma-Aldrich (St. Louis, MO). Furafylline andS-mephenytoin were from Ultrafine Chemicals Ltd. (Manchester, UK). Testosterone was from Fluka (Buchs, Switzerland); tolbutamide was obtained from Sigma/RBI (Natick, MA). Methylp-tolyl sulfide and sulfoxide were from Aldrich Chemical Co. (Milwaukee, WI). 1-benzylimidazole was purchased from Sigma-Aldrich Chemie GmbH (Steinheim, Germany). Na-pyrophosphate, MgCl2, NADPH-sodium salt was obtained from Reanal (Budapest, Hungary).
(±)-Bufuralol hydrochloride, (±)-1′-hydroxybufuralol maleate, CYP2D6- and CYP3A4-specific monoclonal antibodies, CYP2C8/9/19 and NADPH-P450-reductase specific polyclonal antibodies and baculovirus-insect-cell-expressed human enzymes were purchased from BD Gentest (Woburn, MA). Human liver microsomes were from In Vitro Technologies (Baltimore, MD) and from Department of Biochemical Pharmacology, Chemical Research Institute of Hungarian Academy of Sciences (Budapest, Hungary).14C-Dextromethorphan was from Chemical Research Institute of Hungarian Academy of Sciences (Budapest, Hungary). Solvents used for chromatography were of gradient grade and purchased from Merck.
Incubation Conditions.
All incubations were carried out in incubation mixtures containing 6 mM Na-pyrophosphate, 5 mM MgCl2, 5 mM glucose 6-phosphate, 1 U/ml glucose-6-phosphate dehydrogenase in a final volume of 0.5 ml of 0.1 M Tris-HCl buffer, pH 7.4. Tolperisone concentration was 1 and 5 μM (Clint determination and inhibition experiments), or 0.5–1000 μM (enzyme kinetic measurements). In HLM incubations, the mixture contained 1 mg/ml of microsome pool and the reactions were initiated by the addition of 5 mM NADPH and conducted in shaking water bath at 37°C for 20 min. In incubations containing insect cell-expressed recombinant human P450s, the enzyme concentration was 20 pmol/ml (CYP1A2, CYP2A6, CYP2B6, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A4) or 1000 pmol/ml (FMO3). The reactions were initiated by the addition of microsomes and incubated in water bath at 37°C, without shaking, according to the manufacturer's recommendation. Microsomes prepared from wild-type baculovirus-infected cells were used as control. The reaction was stopped with equal volume of ice-cold methanol, centrifuged for 10 min at 7500g, and 10 μl of supernatant was injected into HPLC.
Inhibition Studies with Chemical Inhibitors.
P450 isoform-specific chemical inhibitors or index substrates were used to inhibit tolperisone metabolism in HLM. Furafylline for CYP1A2 (Sesardic et al., 1990; Kunze and Trager, 1993; Tassaneeyakul et al., 1994; Newton et al., 1995), sulfaphenazole for CYP2C9 (Baldwin et al., 1995; Newton et al., 1995), quinidine for CYP2D6 (Newton et al., 1995), ketoconazole for CYP3A4 (Baldwin et al., 1995; Newton et al., 1995; Sai et al., 2000), and S-mephenytoin for CYP2C19 (Goldstein et al., 1994) and CYP2B6 (Küpfer and Preisig, 1984) were used in the concentration range of 1 to 100 μM (quinidine and ketoconazole), 2 to 200 μM (furafylline, sulfaphenazol, S-mephenytoin). Thiourea at 10 mM was used as a competitive inhibitor of FMO3 (Poulsen et al., 1979). SKF-525A (100–1000 μM; Schenkman et al., 1972), 1-aminobenzotriazole (ABT) (1000 μM; Ortiz de Montellano et al., 1984; Xu et al., 1994) and 1-benzylimidazole (BI) (1000 μM, Madan et al., 1993; Grothusen et al., 1996) were used as nonspecific P450 inhibitors to assess the P450-dependent metabolism of tolperisone. All inhibitors were dissolved in methanol and coincubated with tolperisone in the incubation mixture except for furafylline and ABT, which are mechanism-based inhibitors; therefore, those were preincubated with HLM in the presence of NADPH-regenerating system for 10 and 20 min, respectively. In these experiments, the reaction was initiated with tolperisone dissolved in methanol. The methanol concentration in the incubation mixture did not exceed 1% (v/v).
Inhibition of Index Reactions.
Tolperisone was used as inhibitor in the concentration range of 0 to 90 μM—except for dextromethorphan O-demethylase and MpTS oxidase, where tolperisone was 0 to 45 μM and 0 to 1500 μM, respectively—and was coincubated with index substrates. The incubation mixture, quenching, and processing of samples for HPLC measurements were the same as described above.14C-dextromethorphan O-demethylase assay was carried out as previously described (Rodrigues, 1996). Bufuralol hydroxylase assay described by Boobis et al. (1985) was also used as an index reaction of CYP2D6 with modifications. Briefly, 5 to 200 μM of bufuralol was coincubated with 10 to 90 μM tolperisone for 40 min in shaking water bath at 37°C in a total volume of 500 μl. Testosterone 6β-hydroxylase assay was the modification of the method described by Anderson et al. (1995). Testosterone was used in a concentration range of 20 to 500 μM, and the mixture was incubated for 30 min. Tolbutamide hydroxylase was carried out as previously described (Miners et al., 1988) without extraction. Tolbutamide was used in 20 to 500 μM. Phenacetin O-deethylase was carried out as described previously (von Moltke et al., 1996), with slight modifications in the composition of incubation mixture and detection wavelength (249 nm). As index reaction of FMO3, methylp-tolyl-sulfide (MpTS) oxidation was carried out (Pike et al., 1999). Since this reaction is known to be mediated not only by flavin-containing monooxygenases, insect cell-expressed human FMO3 was used. MpTS was added in the concentration range of 10 to 1500 μM. Incubations were performed for 15 min. The produced methylp-tolyl sulfoxide (MpTSO) was monitored with HPLC.
Inhibition of Tolperisone Metabolism with Antibodies.
Before addition to the mixture, pooled HLM were preincubated with the antibodies in triplicates either for 15 min on ice (CYP2D6- and CYP3A4-specific antibodies; 6 and 10 μl of antibodies/100 μg of microsomes, respectively) or for 30 min at room temperature (CYP2C8/9/19-specific and NADPH-P450 reductase-specific antisera; 20 μl and 7.5 μl of antisera/100 μg microsomes, respectively). Anti-reductase inhibition kit contained nonimmune goat serum as control. In experiments with CYP2C8/9/19-specific antiserum, normal goat serum from Invitrogen New Zealand Ltd. (Auckland, NZ) was used as control.
High Performance Liquid Chromatography.
Analytical measurements were carried out using a Merck-Hitachi LaChrom HPLC system equipped with UV detector. Discovery C18 150 × 4.6 mm (5 μm) column with Supelguard 20 × 4 mm (5 μm) column (Supelco, Bellefonte, PA) was maintained at 40°C for all assays.
Tolperisone and hydroxymethyl-tolperisone was monitored at 256 and 251 nm, respectively. The system was operated at 0.8 ml/min flow rate, under isocratic conditions. Mobile phase contained 47% methanol in 0.1 M ammonium-acetate. For the determination of 1′-hydroxybufuralol mobile phases of methanol/0.1 M ammonium-acetate, 27:73 (A) and methanol (B) were used. Gradient (A) was 100 (8 min), 40 (16–17 min), and 100% (20–25 min). Column effluent was monitored at 252 nm, the excitation wavelength of 1′-hydroxybufuralol determined previously (Kronbach et al., 1987). Measurement of 6β-hydroxytestosterone was performed using mobile phases of methanol/0.1 M ammonium-acetate, 45:55 (A) and methanol (B) with a gradient (A) of 100 (4 min), 40 (14–17 min), and 100% (20–26 min). UV detector was set to 254 nm (Anderson et al., 1995). Mobile phases for hydroxytolbutamide analysis were methanol/0.1 M ammonium-acetate, 25:75 (A) and methanol (B). Gradient (A) was 100 (4 min), 70 (10–11 min), and 100% (13–20 min). Detection was carried out at 230 nm (Miners et al., 1988). 4-Acetamidophenol, the CYP1A2 metabolite of phenacetin was also examined using gradient method. Mobile phase A was methanol/0.1 M ammonium-acetate, 5:95 and B was methanol. Gradient (A) was 100 (0 min), 50 (12 min), 100% (14–19 min). Column effluent was monitored by UV detector at 249 nm (UV absorption maximum estimated previously). MpTSO, the metabolite of MpTS was monitored at 233 nm. Mobile phases were methanol/0.1 M ammonium-acetate, 34:66 (A) and methanol (B). Gradient (A) was 100 (5 min), 35 (12 min), and 100% (16–22 min).
LC/MS Measurements.
Merck-Hitachi LaChrom HPLC system was coupled with mass spectrometer (Merck-Hitachi M-8000) with electrospray ionization interface. The enzyme reaction was stopped with equal volume of ice-cold methanol. The samples were kept at −20°C for 30 min. After centrifugation, the supernatant was collected, and the pH was set to 9 and extracted with chlorobutane. Organic phase was evaporated to dryness and redissolved in ethanol, and 10 μl was subjected to HPLC. Eluent A consisted of 140 ml of methanol and 100 ml of water, supplemented with 100 μl 10% formic acid and 200 μl 10% ammonium hydroxide. Eluent B consisted of methanol. Gradient: 0. min: 100% A, 8. min: 60% A, 12. min: 100% A. Flow rate was 0.25 ml/min. Samples were monitored at 255 nm with UV detector. Prontosil C18 ACE EPS 5 μm, 100–2 mm column (Bischoff, Germany) was used at 35°C.
Data Analysis.
Enzyme kinetic data of tolperisone hydroxylation in pooled HLM and recombinant isozymes were fitted to standard Michaelis-Menten plot (reaction rate “V” against substrate concentration “S”) and Eadie-Hofstee plot (“V/S” against “V”). Kinetic parameters (apparent Km andVmax) were calculated using nonlinear regression analysis of GraphPad Prism 2.01 (GraphPad Software Inc., San Diego, CA). Standard deviation and p values of Student'st test were calculated with Microsoft Excel (Redmond, WA). For determination of Ki, “Km,app” method was used (Kakkar et al., 1999). Clint in pooled HLM was calculated from initial reaction rate using the following equation:
Results
Intrinsic Clearance of Tolperisone.
The in vitro Clint of tolperisone was determined using individual microsomes from nine donors (three female and six male). Tolperisone had a Clint of 1.27 ± 0.16 ml/min/g of liver, determined as the mean of nine individual clearance values ± S.D. The female and the male pool resulted in 1.39 ± 0.22 and 1.15 ± 0.20 ml/min/g of liver, respectively (mean of three and six individual samples ± S.D., respectively). The sex difference in Clint of the compound was not statistically significant. The metabolism was NADPH-dependent, since the omission of this coenzyme completely abolished the consumption of tolperisone (data not shown).
Tolperisone is metabolized mainly to hydroxymethyl-tolperisone (hydroxylation on methyl group of 4-methyl-phenyl moiety) in HLM that can be monitored with UV detector. LC/MS measurements revealed three main metabolites represented as molecular ions at m/z 261, 263, and 247. Metabolites m/z 261 (M1) and m/z263 (M2) were identified as hydroxymethyl-tolperisone and carbonyl-reduced form of M1, respectively. Both metabolites were also found in 24-h rat urine (Miyazaki et al., 1972).
Isoform-Specific Chemical Inhibitors/Index Substrates.
P450 isoform-specific chemical inhibitors or index substrates (i.e., competitive inhibitors) were used to inhibit the consumption of tolperisone (5 μM) and the formation of M1 metabolite in pooled HLM. Furafylline (CYP1A2), sulfaphenazole (CYP2C9), S-mephenytoin (CYP2C19), quinidine (CYP2D6), and ketoconazole (CYP3A4) were tested as inhibitors. The summarized results are shown in Table1. The only compound found to substantially inhibit tolperisone consumption and M1 formation was quinidine at a diagnostic 10 μM concentration. Ketoconazole at 10 μM inhibited the loss of parent compound and M1 formation to a lesser extent. S-mephenytoin and sulfaphenazole resulted in slight and concentration independent (data not shown) inhibition both in consumption and M1 formation. Thiourea was also used to assess the contribution of FMO3 to the metabolism of tolperisone. No inhibition was seen at 10 mM (data not shown).
Inhibition of tolperisone consumption and M1 formation with chemical inhibitors
Inhibitory Antibodies.
Human CYP2D6 and human CYP3A4-specific monoclonal and human CYP2C8/9/19-specific polyclonal antibodies were used to test the contribution of respective P450 enzymes to the in vitro metabolism of tolperisone (5 and 50 μM) in pooled human liver microsomes. CYP2D6- and CYP2C-specific antibodies inhibited the clearance of tolperisone and M1 formation (Table 2); antibodies against CYP3A4 had no effect (data not shown). Anti-CYP2C-inhibited tolperisone clearance more effectively when 50 μM substrate was used, whereas the inhibitory effect of anti-CYP2D6 decreased. The same pattern was seen in M1 formation, the 70% inhibition of anti-CYP2D6 at 5 μM decreased to about 50% at 50 μM. These results point toward the decreasing relative contribution of CYP2D6 along with increasing tolperisone concentration. Although CYP2C-specific antibodies resulted in significant inhibition, these results were difficult to interpret since nonimmune goat serum had an intrinsic, yet unidentified metabolizing capacity.
Inhibition of tolperisone clearance and M1 formation with antibodies in HLM
Recombinant Enzymes.
Insect cell-expressed recombinant human P450s (CYP1A2, CYP2A6, CYP2B6, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A4) were used to study the contribution of these enzymes to the metabolism of tolperisone. The reaction mixture contained 20 pmol/ml of each P450 and 1 μM tolperisone. Under these conditions, CYP2D6 and CYP2C19 containing microsomes metabolized tolperisone substantially to the main M1 metabolite (Fig. 1). Recombinant CYP1A2 also catalyzed tolperisone hydroxylation at an order of magnitude lower rate than CYP2D6 and CYP2C19. CYP2B6 also contributes to the consumption of parent compound, however metabolite produced by this isozyme still needs to be identified.
Comparison of tolperisone-consuming and M1-producing activity of baculovirus-infected insect cell-expressed P450s.
Twenty picomoles per milliliter of each enzyme was used at tolperisone concentration of 1 μM. After 20 min of incubation, the reaction was stopped, and unchanged parent compound and produced M1 were determined with HPLC. The amount of consumed tolperisone was calculated, and the data were converted into activity. Each bar represents the mean of a triplicate measurement ± S.D. A, comparison of tolperisone-metabolizing activity of expressed human P450s. B, comparison of M1 formation rates of expressed human P450s.
Since aforementioned results implied that NADPH-dependent, non-P450 enzymes might also contribute to the metabolism of tolperisone, inhibition of MpTS oxidation, as an FMO3-mediated reaction, was carried out. This reaction is known to be nonspecific for FMO3 (i.e., P450s are also involved, Pike et al., 1999), therefore we used recombinant, insect cell-expressed human FMO3 containing microsomes. Analysis of the data revealed a Ki of 1197 μM in MpTS oxidase inhibition. The consumption of tolperisone was examined in rFMO3 containing 1 nmol enzyme/ml. In this latter experiment rFMO3 resulted in a turnover number/specific activity of 60 pmol/min/nmol enzyme.
Kinetics of Tolperisone Hydroxylation.
Enzyme kinetic analysis of M1 formation was carried out to study the main metabolic route of tolperisone biotransformation. In experiments using HLM tolperisone was used in the concentration range of 1 to 1000 μM. Data of M1 formation were fitted to Michaelis-Menten kinetics (Fig. 2). Calculated apparent kinetic values were summarized in Table 3. Kinetic analysis of M1 formation showed biphasic kinetics, suggesting the contribution of at least two populations of P450s. Eadie-Hofstee plot (Fig. 2B) of data points also revealed that there is a highKm and a lowKm component of M1 formation. Fine characterization of M1 formation in expressed human P450s revealed CYP2D6 as the low Km enzyme of methyl-hydroxylation (Table 4). CYP2C19 showed medium Km and CYP1A2 turned out to be the high Km component. The apparent Vmax rank order was CYP2D6 < CYP1A2 < CYP2C19. The Clint(calculated asVmax/Km) implied that CYP2D6 played the essential role in tolperisone metabolism.
Kinetic analysis of tolperisone metabolism in HLM.
A, Michaelis-Menten plot of M1 formation reaction. The dotted line represents one-Km, the solid line represents two-Km kinetic calculation. The corresponding r2 values are 0.95 and 0.998, respectively. B, Eadie-Hofstee plot of M1 metabolite formation in HLM.V and S stand for the reaction velocity and tolperisone concentration, respectively. Tolperisone was used at 1 to 1000 μM.
Enzyme kinetic parameters of tolperisone hydroxylation in HLM
Kinetic parameters of tolperisone hydroxylation in baculovirus-infected insect cell expressed human P450s
Inhibition Studies for Ki Determination.
To determine inhibitory potential of tolperisone toward the prominent isozyme, compound was tested to inhibit dextromethorphanO-demethylation and bufuralol hydroxylation—widely used index reactions of CYP2D6. Enzyme kinetic analysis of the data with the “Km,app” method (Kakkar et al., 1999) revealed that tolperisone inhibited dextromethorphanO-demethylation and bufuralol-hydroxylation with theKi of 17 and 30 μM, respectively.
Tolperisone was also used as inhibitor in testosterone 6β-hydroxylation, phenacetin O-deethylation and tolbutamide-hydroxylation, the index reactions of CYP3A4, CYP1A2, and CYP2C9, respectively. Tolperisone did not inhibit these index reactions in the concentration range used for inhibition of CYP2D6 index reaction (data not shown).
Contribution of Microsomal Non-P450 Enzymes.
To test whether non-P450, NADPH-dependent enzymes contribute to the in vitro metabolism of tolperisone in HLM, nonspecific chemical P450 inhibitors (SKF-525A, 1-aminobenzotriazole, 1-benzylimidazole) and anti-NADPH-P450 reductase polyclonal antibodies were examined to inhibit P450-dependent reactions. Results are shown in Table 1(SKF-525A, ABT, and BI).
SKF-525A at 1000 μM completely blocked, at 100 μM inhibited by 79% of M1 formation in the incubation mixture containing 5 μM tolperisone. Contrary, the clearance was not inhibited to such a large extent, 100 and 1000 μM SKF-525A inhibited by 39 and 61%, respectively. The inhibitory pattern of ABT and BI were the same as that of SKF-525A; parent loss was inhibited by 43–61%, while M1 formation was completely abolished.
Anti-NADPH-P450 reductase antibodies reduced tolperisone loss by 43%. This inhibitory effect is significantly larger toward M1 metabolite formation (76%). LC/MS measurements revealed that P450-reductase-specific antibodies inhibited the formation of M1 and M2 significantly, but formation of m/z 247 was not affected (data not shown).
Discussion
In the present study the in vitro pathways of tolperisone metabolism was examined in pooled human liver microsomes and in recombinant enzymes. In previous pharmacokinetic studies it was found that tolperisone had a low bioavailability (20%, Miskolczi et al., 1987). Here we demonstrated that the rate of NADPH-dependent Clint of tolperisone could account for the low in vivo bioavailability.
Experiments studying metabolic stability of tolperisone also revealed that the compound is metabolized mainly to hydroxymethyl-tolperisone (M1), in vitro. Stochiometric calculations predicted the formation of other, substantial, non-UV active metabolites besides the main M1. LC/MS measurements showed that as well as M1 (m/z 261), the carbonyl-reduced form of this metabolite (M2, m/z 263), and another metabolite that is two mass units greater than parent compound was produced (m/z 247). Since several carbonyl-reduced metabolites were identified previously in rats (Miyazaki et al., 1972), and one of these was identified in HLM in the present study (M2), it was reasonable to assume that metabolite at m/z 247 was identical with the carbonyl-reduced parent compound. Nonspecific P450 inhibitors (SKF-525A, ABT, BI, and NADPH-P450 reductase-specific antibodies) potently inhibited M1 formation, however parent compound consumption was inhibited to a much lesser extent (40–60% residual microsomal activity at 100% P450 inhibition). This suggested the substantial contribution of P450-independent enzymes. Additionally, anti-reductase antibodies did not inhibit the formation of metabolitem/z 247, whereas the amount of M1 and M2 was decreased. These indirect evidences implied that besides P450s, a significant P450-independent route is involved in tolperisone metabolism that is assumed a microsomal carbonyl reductase. However, further supportive results are needed to identify the reductase and evaluate its role in tolperisone metabolism.
The P450-dependent metabolic processes were characterized in details. In our hands, the most potent chemical inhibitor of tolperisone consumption and M1 metabolite formation was quinidine that implied the main role of CYP2D6. This result was in concert with results of recombinant P450s, where the most active isoform (i.e., the enzyme with the highest turnover number) was CYP2D6 both in parent loss and M1 formation. More supportive data of the key role of CYP2D6 came from the experiments with specific inhibitory antibodies; 70% inhibition of M1 formation meant that the majority of the P450-dependent biotransformation is mediated by CYP2D6. S-mephenytoin inhibited parent loss about to the same extent as quinidine (22%), however the effect to M1 formation was weak (16%). On the other hand, the activity of recombinant CYP2C19 revealed 60% of rCYP2D6 activity in both compound loss and M1 formation placing recombinant CYP2C19 to the second most active P450 in tolperisone metabolism. Despite this high activity in expressed enzyme system, we concluded that at “physiological” tolperisone concentration the role of CYP2C19 is less essential than that of CYP2D6. The results given by inhibitory antibodies are controversial, since an unidentified interaction of tolperisone with goat serum was seen. However, it was clearly shown that at a higher concentration, the relative contribution of CYP2D6 decreased. This finding was supported with the results of enzyme kinetic characterization of tolperisone hydroxylation. Recombinant CYP2D6 revealed the low Km enzyme with low apparent Vmax indicating that at low concentration CYP2D6 plays the prominent role, whereas increasing the concentration of tolperisone increased the role of CYP2C19 as a medium Km-highVmax enzyme. CYP1A2 turned out to be the highest Km counterpart; however, because of the relative abundance of this isozyme its role is not negligible. Moreover, the role of a nonpolymorphic enzyme is important mainly in a subject of poor metabolizer phenotype both for CYP2D6 and CYP2C19. Calculation of Clint(Vmax/Km) revealed a rank order (CYP2D6 > CYP2C19 > CYP1A2) that assumed to be a valid order of activity for microsomal metabolism, however accurate scaling from heterologously expressed enzymes to human liver microsomes requires additional factors to consider (Venkatakrishnan et al., 2000; Nakajima et al., 2002).
Furafylline, a potent mechanism-based inactivator of CYP1A2 (Kunze and Trager, 1993) did not inhibit strongly tolperisone consumption, however the 25% inhibition in M1 formation may represent its ratio in the contribution to hydroxylation reaction as demonstrated by recombinant P450s. The inhibition measured using ketoconazole and sulfaphenazole was not strengthened by other methods; experiments with recombinant P450s and inhibitory antibodies did not support the contribution of CYP3A4 and CYP2C9. The effect is probably due to the fact that in this concentration range ketoconazole and sulfaphenazole are no longer selective for CYP3A4 and CYP2C9, respectively (Newton et al., 1995;Moody et al., 1999).
In our hands, tolperisone revealed to be a weak inhibitor of FMO3 index reaction in recombinant enzyme. The measuredKi, the small turnover number, and the inability of thiourea to inhibit tolperisone metabolism in HLM implied that this enzyme is not likely to contribute substantially to the metabolism of tolperisone, in vivo.
In the present study, it has been demonstrated that both P450-dependent and P450-independent microsomal biotransformations are involved in tolperisone metabolism, in vitro. Proposed metabolic routes are summarized in Fig. 3. Hydroxymethyl metabolite formation revealed to be the main P450-mediated metabolic pathway. CYP2D6 was identified as the key enzyme in metabolism through M1 formation and involvement of CYP2C19 and CYP1A2 were also shown. It was evidenced that P450-independent metabolism was mediated to a small extent by FMO3. Metabolites detected and indirect evidences from inhibition studies pointed toward the substantial involvement of presumable microsomal carbonyl reductase in the metabolism of tolperisone.
Proposed in vitro metabolic pathways of tolperisone.
Dashed arrows indicate the possible ways of M2 formation. Gray letters are used for the assumed enzymatic processes and metabolites.
Acknowledgments
The authors thank Gabriella Pásztor for help in metabolite identification and Teréz Merkl and Marianna Borsos for skilful technical assistance.
Footnotes
- Abbreviations used are::
- AUC
- area under the curve
- P450
- cytochrome P450
- FMO
- flavin-containing monooxygenase
- HPLC
- high-performance liquid chromatography
- HLM
- human liver microsomes
- SKF-525A
- proadifen-hydrochloride
- ABT
- 1-aminobenzotriazole
- BI
- 1-benzylimidazole
- MpTS
- methylp-tolyl-sulfide
- MpTSO
- methyl p-tolyl sulfoxide
- LC/MS
- liquid chromatography-mass spectrometry
- V
- reaction rate
- S
- substrate concentration
- M1
- hydroxymethyl-tolperisone
- M2
- carbonyl-reduced form of hydroxymethyl-tolperisone
- Clint
- intrinsic clearance
- Received October 28, 2002.
- Accepted February 10, 2003.
- The American Society for Pharmacology and Experimental Therapeutics