scispace - formally typeset
Search or ask a question

Showing papers in "Drug Metabolism and Disposition in 1996"


Journal Article
TL;DR: The large interindividual variabilities in cotinine formation and immunochemically determined CYP2A6 levels were observed in human liver microsomes, suggesting genetic polymorphism of CYP 2A6.
Abstract: Nicotine is primarily metabolized to cotinine in humans. In this study, human cytochrome P450 (CYP) isoform involved in cotinine formation was identified. The formation of cotinine in 16 human liver microsomes was determined with a 50 microM nicotine concentration and with a cytosol preparation as a source of aldehyde oxidase. Cotinine formation in human liver microsomes significantly correlated with immunochemically determined CYP2A6 levels (r = 0.663, p < 0.05), coumarin 7-hydroxylase activities (r = 0.831, p < 0.01), and cotinine 3'-hydroxylase activities (r = 0.735, p < 0.01) that are responsible for CYP2A6. In inhibition studies, cotinine formation in human liver microsomes was inhibited by coumarin and rabbit anti-rat CYP2A1 antibody specifically. When the capability of microsomes of B-lymphoblastoid cells expressing human CYPs to perform biotransformation of nicotine to cotinine was determined, cDNA-expressed CYP2A6 exhibited the highest cotinine formation. The KMapp values from microsome expressing CYP2A6 cDNA were similar to the value obtained from human liver microsomes. The large interindividual variabilities in cotinine formation and immunochemically determined CYP2A6 levels were observed in human liver microsomes, suggesting genetic polymorphism of CYP2A6. Nicotine is a new in vivo probe for phenotyping of CYP2A6 in humans.

429 citations


Journal Article
TL;DR: Results suggest that in human fetal livers at least two P450 enzymes, a form of P450 that is immunoreactive P4501A1 and P4503A7, are actually expressed and these enzymes are suggested as being involved in the activation of the (+)- and (-)-enantiomers of 7,8-dihydroxy-7, 8- dihydrobenzo[a]pyrene and the carcinogenic mycotoxins.
Abstract: Levels and catalytic activities of cytochrome P450 (P450) enzymes involved in the oxidation of drugs and carcinogens were determined in human adult lungs and fetal livers and compared with those in microsomes from adult livers. P450s immunoreactive with anti-human P4501A1 and anti-human P4503A antibodies were detected in fetal liver microsomes by immunoblotting analysis, and P450s related P4501A1, 2A6, 2C9, 2E1, and 3A4 were determined in adult lung microsomes; all of these P450 enzymes were detected in much higher amounts in adult liver microsomes except that P4501A2 was only the 1A subfamily of P450 found in adult livers. Drug oxidation activities with the substrates ethoxyresorufin, coumarin, 7-ethoxycoumarin, bufuralol, and testosterone were determined in these microsomes, and we found that none of the activities were higher in microsomes of adult lungs and fetal livers than in adult livers. Activation of procarcinogens to reactive metabolites that induce umu gene expression in Salmonella typhimurium TA1535/pSK1002 or NM2009 was also examined and it was found that activities with (+)- and (-)-enantiomers of 7,8-dihydroxy-7,8-dihydrobenzo[a]pyrene were higher in fetal liver microsomes than adult lung or liver microsomes. The adult liver and lung activities for these two procarcinogens were similar on the basis of microsomal protein contents despite the fact that p450 contents are higher in liver than lung microsomes. alpha-Naphthoflavone, a known inhibitor of P4501A-related activities, did not affect these procarcinogen activation in fetal liver microsomes. Fetal liver microsomes catalyzed activation of aflatoxin B1 and sterigmatocystin, two procarcinogens known to be activated by P4503A4/7 in humans, although activation of carcinogenic arylamines that are good substrates for P4501A2 was much lower in microsomes of fetal livers and adult lungs than in adult livers. These results suggest that in human fetal livers at least two P450 enzymes, a form of P450 that is immunoreactive P4501A1 and P4503A7, are actually expressed and these enzymes are suggested as being involved in the activation of the (+)- and (-)-enantiomers of 7,8-dihydroxy-7,8-dihydrobenzo[a]pyrene and the carcinogenic mycotoxins, respectively. The exact nature of the former enzyme in fetal livers is unknown. In adult human lungs, several P450 enzymes are expressed, although the precise roles of these enzymes in the oxidation of xenobiotics were not determined due to the low level of expression of these P450s.

256 citations


Journal Article
TL;DR: 6',7'-Dihydroxybergamottin is a potent inhibitor of CYP3A activity, accounts for the difference in inhibition between grapefruit Juice and orange juice in vitro, and may be primarily responsible for the effects of grapefruit juice on cytochrome P450 activity in humans.
Abstract: Grapefruit juice inhibits the metabolism of substrates for enzymes of the CYP3A subfamily. However, the identity of the inhibitor has not been established. Grapefruit juice was extracted into methylene chloride and chromatographed by HPLC, and the effect of the HPLC eluent on CYP3A activity was assessed by measuring 6beta-hydroxytestosterone formation in rat liver microsomes. Significant inhibition was associated with a fraction of HPLC eluent containing a single peak, with a retention time of 16 min. The substance producing this peak was isolated using TLC and identified, using NMR and MS, as 6',7'-dihydroxybergamottin (C21H2406; molecular weight, 372), a furanocoumarin (psoralen) compound. The concentration of 6',7'-dihydroxybergamottin required to inhibit 6beta-hydroxytestosterone formation by 50% was 25 microM. Grapefruit juice reduced CYP3A activity to a significantly greater extent (p < 0.05) than did orange juice, which contained no measurable 6',7'-dihydroxybergamottin (28.6% vs. 62.2% of control activity). The addition of 6',7'-dihydroxybergamottin (30 microM) to orange juice decreased CYP3A activity to values comparable to those observed with grapefruit juice. 6',7'-Dihydroxybergamottin is a potent inhibitor of CYP3A activity, accounts for the difference in inhibition between grapefruit juice and orange juice in vitro, and may be primarily responsible for the effects of grapefruit juice on cytochrome P450 activity in humans.

222 citations


Journal Article
TL;DR: Overall, the results demonstrated that both the preparations of small intestines and Caco-2 cells exhibited significant drug-metabolizing enzyme activities, although several differences were noted between the intestinal enzymes in the animals or in the Caco -2 cells and those found in humans.
Abstract: Drug-metabolizing enzymes were studied in subcellular fractions of dog, monkey, and human small intestines, and in the human adenocarcinoma cell line Caco-2, a commonly used in vitro absorption model. Immunoblot analysis indicated the presence of enzymes related to cytochrome P450 (CYP) 1A1/CYP1A2, CYP2D6, CYP3A, and carboxylesterases (ESs) in human and monkey intestines, and of CYP3A and ES in dog intestines. Catalytically, human and monkey intestines exhibited significant and comparable testosterone 6 beta-hydroxylase, (+)-bufuralol 1'-hydroxylase, and ES activities. In contrast, dog intestine possessed moderate testosterone 6 beta-hydroxylase, much lower ES, and undetectable bufuralol hydroxylase activities. In addition, low tolbutamide methylhydroxylase activity was observed in human and monkey intestines, but not in dog intestines. Of the phase I enzymes investigated, only ES was detected immunologically and functionally in Caco-2 cells. With respect to phase II enzymes, human and monkey intestines contained relatively high intestinal glucuronyltransferase, N-acetyltransferase (NAT), sulfotransferase, and glutathione S-transferase activities. Except for NAT, all phase II enzymes studied were detectable in dog intestines. In Caco-2 cells, acetaminophen sulfation activity was below the limit of detection, whereas all other conjugating activities were evident. Studies of enzyme kinetics and inhibition by known inhibitors of testosterone 6 beta-hydroxylase activity, the major intestinal mono-oxygenase in all species, revealed some similarities between the responsible enzymes. Comparative studies with human liver microsomes suggested the possible involvement of CYP3A enzymes in the intestinal catalysis of testosterone 6 beta-hydroxylation similar to those observed with human hepatic CYP3A. Further studies on ESs, however, revealed multiplicity and species and/or tissue differences in the microsomal and cytosolic enzymes. Based on kinetic studies, monkey intestines and Caco-2 cells possessed NAT activities, with properties similar to those in human intestine and liver. Overall, the results demonstrated that both the preparations of small intestines and Caco-2 cells exhibited significant drug-metabolizing enzyme activities, although several differences were noted between the intestinal enzymes in the animals or in the Caco-2 cells and those found in humans.

214 citations


Journal Article
TL;DR: The results suggest that human UDP-glucuronosyltransferase 1.4 may be an important enzyme for the detoxication of environmentally derived amines and sapogenins and for the conjugation of progestins.
Abstract: Glucuronide conjugation of tertiary amine xenobiotics represents a unique and important metabolic pathway for these compounds in humans. In this study, we show that human UDP-glucuronosyltransferase 1.4 protein, stably expressed in human embryonic kidney 293 cells, catalyzes the N-glucuronidation of primary, secondary, and tertiary amine substrates. In addition, the substrate specificity of the expressed enzyme toward many hydroxylated and carboxylic acid-containing compounds was examined. Of the hydroxylated compounds tested, only sapogenins gave glucuronidation rates comparable with those observed for amine substrates. The apparent KM and Vmax values for sapogenins were such that the efficiency of glucuronidation (Vmax/KM) for these compounds was higher than that determined for amine substrates. Human UDP-glucuronosyltranferase 1.4 also catalyzes the glucuronidation of monoterpenoid alcohols and simple phenolic compounds. The enzyme kinetic values determined for these substrates suggested that this enzyme may have relatively limited significance for the conjugation of these classes of compounds. Of the endobiotics tested, androstanediol and progestins were glucuronidated at high rates by expressed human UDP-glucuronosyltransferase 1.4 protein. The glucuronidation efficiency for 5alpha-pregnane-3beta,20alpha-diol was comparable with that determined for the sapogenins. Because UDP-glucuronosyltransferases are integral membrane proteins, the effects of different detergents on the catalytic activity of the expressed enzyme were determined. The results show that detergents (such as Lubrol PX, Emulgen 911, and Triton X-100) are inhibitory for the quaternary ammonium-linked glucuronidation of chlorpromazine and imipramine catalyzed by expressed human UDP-glucuronosyltransferase 1.4. In contrast, CHAPS and nonanoyl-N-methylglucamide are less inhibitory toward the glucuronidation of these compounds. The results suggest that human UDP-glucuronosyltransferase 1.4 may be an important enzyme for the detoxication of environmentally derived amines and sapogenins and for the conjugation of progestins.

204 citations


Journal Article
A Ghosal1, H Satoh, P E Thomas, E Bush, D Moore 
TL;DR: Inhibition studies showed that MDZ hydroxylation was strongly inhibited by ketoconazole and Ro 23-7637 in rat, human, and cDNA-expressed human CYP3A4 microsomes, thus suggesting that members of the CYP 3A4 subfamily were involved in the metabolism.
Abstract: Midazolam (MDZ) is metabolized in human liver microsomes by the cytochrome P450 (CYP) 3A subfamily to 1'-hydroxy (1'-OH) and 4-hydroxy (4-OH) metabolites. MDZ is metabolized in the rat primarily to 4-OH MDZ, 1'-OH MDZ, and 1',4-dihydroxy (1',4-diOH) MDZ. The kinetics of 4-OH and 1'-OH metabolite formation were determined using hepatic microsomes from control, Ro 23-7637 and dexamethasone-treated male rats. KM values for the major metabolite, 4-OH MDZ, were 24.5, 43.1, and 32.8 microM, and the corresponding Vmax values were 5.9, 28.9, and 13 nmol/mg/min for the control, DEX, and Ro 23-7637-treated animals, respectively KM values for 1'-hydroxylation of MDZ (the major metabolite) after incubation with human liver microsomes from three individuals were 5.57, 2.50, and 3.56 microM, and the corresponding Vmax values were 4.38, 0.49, and 0.19 nmol/mg/min, respectively. In parallel studies using cDNA-expressed human CYP3A4 microsomes, the KM for 1'-OH formation was 1.56 microM, and the corresponding Vmax was 0.16 nmol/mg/min. MDZ was not metabolized by cDNA-expressed human CYP2D6, CYP2E1, or CYP1A2, thus confirming that these isoforms were not responsible for its biotransformation. The formation of 1',4-diOH metabolite in rat and 1'-OH formation in cDNA-expressed human CYP3A4 microsomes showed a decrease in velocity at high substrate concentrations. Inhibition studies showed that MDZ hydroxylation was strongly inhibited by ketoconazole and Ro 23-7637 in rat, human, and cDNA-expressed human CYP3A4 microsomes. alpha-Naphthoflavone stimulated 1'-OH metabolite formation in human and cDNA-expressed human CYP3A4 microsomes at low concentration (10 microM). Naringenin, a flavonoid present in grapefruit juice, also inhibited MDZ metabolism in human liver microsomes. Immunoinhibition studies revealed that polyclonal anti-rat CYP3A2 antibody inhibited MDZ metabolism 80-90% in rat, human, and cDNA-expressed human CYP3A4 microsomes, thus suggesting that members of the CYP3A4 subfamily were involved in the metabolism.

202 citations



Journal Article
TL;DR: Examples of each of the xenobiotic-metabolizing CYP1, CYP2, and CYP3 subfamilies were detected in low levels in human normal breast tissue and tumors, indicating the machinery for possible in situ bioactivation of xenobiotics and modification of therapeutic drugs is thus present in human breast tissue.
Abstract: In an effort to determine which members of the cytochrome P450 (CYP) superfamily are expressed in human breast tissue and tumors, RNA-polymerase chain reaction studies have been undertaken. Detection of expressed CYP mRNAs identifies those forms of the enzyme that are capable of expression in breast tissue, and provides insight into the potential for in situ xenobiotic and therapeutic drug metabolism. CYP1A1 mRNA was present in (5/11) breast tissues and (6/13) tumors. When normal and tumor tissues were from the same individuals, higher amplification occurred in normal tissues. CYP1B1 mRNA was present in all but one tissue, and CYP2C mRNA forms were present in all of the tissues. CYP3A4 mRNA was present in (8/11) normal breast tissues and (2/13) tumor tissues, and CYP3A5 mRNA was present in (9/11) normal tissues and (2/13) tumor tissues. The expression of the CYP3A mRNA forms was not coincident, suggesting differential regulation. CYP2D6 mRNA was present in (10/11) normal breast tissue and (10/13) tumors. Two splice variants of CYP2D6 mRNA were also detected; one with a 207 bp intron spliced in was detected in all of the normal tissue samples and (11/13) tumors, whereas another (which lacks a 3'-portion of exon 6) was detected in (9/11) normal breast tissues and (7/13) tumors. Thus, examples of each of the xenobiotic-metabolizing CYP1, CYP2, and CYP3 subfamilies were detected in low levels in human normal breast tissue and tumors. The machinery for possible in situ bioactivation of xenobiotics and modification of therapeutic drugs is thus present in human breast tissue.

182 citations


Journal Article
N Holtbecker1, M F Fromm, Heyo K. Kroemer, E E Ohnhaus, H Heidemann 
TL;DR: It is concluded that there is a relevant interaction between nifedipine and rifampin, and the reduction of nifingipine bioavailability during enzyme induction is most likely due to rifampsin-induced gut wall metabolism.
Abstract: The calcium channel blocker nifedipine is metabolized by cytochrome P450 3A4, which is present in liver and mucosa of the small bowel. Cytochrome P450 3A4 is inducible by the tuberculostatic rifampin in liver and the small bowel. The contribution of gut wall metabolism to total clearance of nifedipine before and during induction has not been determined in detail. We therefore investigated the nifedipine-rifampin interaction, with special emphasis on the contribution of gut wall metabolism to total metabolism of nifedipine before and during administration of rifampin. Pharmacokinetics of nifedipine were studied in six healthy volunteers on separate days by administration of 20 micrograms/kg body weight nifedipine iv and 20 mg nifedipine orally before and after 7 days of rifampin treatment (600 mg/day). Enzyme induction did not significantly alter pharmacokinetics of nifedipine after iv administration. In contrast, oral clearance of nifedipine increased from 1.5 +/- 0.2 liters/min to 20.9 +/- 8.3 liters/min (p < 0.01) and bioavailability decreased from 41.2 +/- 5.4% to 5.3 +/- 2.7% (p < 0.001). Although hepatic extraction of nifedipine was not significantly altered during induction (47.4 +/- 6.6% versus 67.4 +/- 20.2%; ns), the calculated extraction of nifedipine in gut wall mucosa increased from 21.8 +/- 13.3% to 75.8 +/- 28.2% (p < 0.05). We conclude that there is a relevant interaction between nifedipine and rifampin. The reduction of nifedipine bioavailability during enzyme induction is most likely due to rifampin-induced gut wall metabolism.

173 citations


Journal Article
TL;DR: The results strongly support the hypothesis that metabolically based in vivo drug interactions may be predicted from human in vitro microsomal data and suggest that fluconazole can be expected to interact with any drug whose clearance is dominated by P450s 2C9, 3A4, and other as yet undefined isoforms.
Abstract: Consistent with expectations based on human in vitro microsomal experiments, administration of fluconazole (400 mg/day) for 6 days to six human volunteers significantly reduced the cytochrome P450 (P450)-dependent metabolic clearance of the warfarin enantiomers. In particular, P4502C9 catalyzed 6- and 7-hydroxylation of (S)-warfarin, the pathway primarily responsible for termination of warfarin's anticoagulant effect, was inhibited by approximately 70%. The change in (S)-warfarin pharmacokinetics caused by fluconazole dramatically increased the magnitude and duration of warfarin's hypoprothrombinemic effect. These observations indicate that co-administration of fluconazole and warfarin will result in a clinically significant metabolically based interaction The major P450-dependent, in vivo pathways of (R)-warfarin clearance were also strongly inhibited by fluconazole. 10-Hydroxylation, a metabolic pathway catalyzed exclusively by P4503A4, was inhibited by 45% whereas 6-, 7-, and 8-hydroxylations were inhibited by 61, 73, and 88%, respectively. The potent inhibition of the phenolic metabolites suggests that enzymes other than P4501A2 (weakly inhibited by fluconazole in vitro) are primarily responsible for the formation of these metabolites in vivo as predicted from in vitro kinetic studies. These data suggest that fluconazole can be expected to interact with any drug whose clearance is dominated by P450s 2C9, 3A4, and other as yet undefined isoforms. Overall, the results strongly support the hypothesis that metabolically based in vivo drug interactions may be predicted from human in vitro microsomal data.

169 citations


Journal Article
TL;DR: The prediction generated from these studies, that fluconazole is a potent in vivo inhibitor of warfarin metabolism, is tested in complementary studies reported in the accompanying article, "Warfarin-Fluconazoles II".
Abstract: The antifungal agent fluconazole was found to be a potent inhibitor of cytochrome P450 (P450) 2C9 (Ki = 7-8 microM), the principal enzyme responsible for the clearance (85%) of the more potent anticoagulant (S)-warfarin to the inactive (S)-7- and (S)-6-hydroxywarfarin metabolites in vivo. Fluconazole was also found to be a potent inhibitor of the P4503A4-catalyzed formation of (R)-10-hydroxywarfarin (Ki = 15-18 microM) as well as the low KM P450 enzymes responsible for the formation of (R)-6-, (R)-7-, and (R)-8-hydroxywarfarin (Ki = 2-6 microM). By contrast, experiments with the P4501A2 inhibitor furafylline and cDNA-expressed P4501A2 indicate that fluconazole is a weak inhibitor of this enzyme (Ki > 800 microM), as measured by the inability of fluconazole to significantly suppress the P4501A2-dependent 6-hydroxylation of (R)-warfarin. The prediction generated from these studies, that fluconazole is a potent in vivo inhibitor of warfarin metabolism, , is tested in complementary studies reported in the accompanying article, "Warfarin-Fluconazole II".

Journal Article
TL;DR: It is indicated that CYP3A4 is the major catalyst involved in fentanyl oxidation to norfentanyl in human liver, which could lead to marked perturbations in fentanyl disposition and, hence, analgesic response.
Abstract: The microsomal metabolism of fentanyl, a synthetic opioid commonly used in anesthesia, was investigated in human liver. Incubation of fentanyl with human hepatic microsomes fortified with NADPH resulted in the formation of a single major metabolite, namely norfentanyl, as determined by GC/MS. No evidence was obtained for the formation of either desproprionylfentanyl or N-phenylpropionamide, the latter arising via N-dealkylation of the fentanyl amide nitrogen. Kinetic analysis of microsomal fentanyl oxidation revealed a single K(m) of 117 microM and a Vmax of 3.86 nmol of norfentanyl formed/min/nmol of cytochrome P450 (P450). Studies using chemical inhibitors of human P450 enzymes revealed that only agents known to inhibit CYP3A4 (e.g. ketoconazole and erythromycin) were capable of strongly inhibiting (> or = 90%) microsomal fentanyl oxidation. Marked inhibition (> 90%) of norfentanyl formation by liver microsomes was also observed with polyclonal antibodies to CYP3A4, whereas antibodies to other human P450s were without effect. Furthermore, rates of norfentanyl production by 10 individual human liver samples were highly correlated (r2 = 0.876, F = 56.46 p < 0.001) with immunochemically determined levels of CYP3A4 present in the samples but not with levels of CYP2C8, CYP2C9, CYP2C19, or CYP2E1. Our results indicate that CYP3A4 is the major catalyst involved in fentanyl oxidation to norfentanyl in human liver. Alterations in CYP3A4 levels or activity, as well as the concomitant administration of other therapeutic agents metabolized by this P450 enzyme, could lead to marked perturbations in fentanyl disposition and, hence, analgesic response.

Journal Article
TL;DR: Caco-2 cells serve as an excellent model to dissect the relative roles played by p-glycoprotein-mediated efflux and CYP3A-catalyzed oxidation in modulating the overall absorption of CsA and other such compounds.
Abstract: Transport of cyclosporin A (CsA) across Caco-2 cells is modulated by its directional efflux, mediated by a p-glycoprotein-like pump (Augustijns et al., Biochem. Biophys. Res. Comm. 197:360-365, 1994). In addition to this unidirectional flux, oxidative metabolism of CsA by cytochrome P450 is likely to influence the absorption of this cyclic peptide across intestinal mucosa. Thus, metabolism of CsA in the in vitro Caco-2 cell culture system was investigated. Formation of several metabolites was observed during the course of CsA transport across Caco-2 cell monolayers. Results from LC/MS/MS experiments revealed that the major metabolite was 1eta-hydroxy CsA (M-17), one of the three major metabolites produced by CYP3A4 present in both the liver and small intestine in humans. Preincubation of Caco-2 cell monolayers with troleandomycin, a specific inhibitor for the microsomal CYP3A protein, reduced the formation of the metabolite M-17, suggesting that an enzyme that functionally resembles CYP3A is responsible for the formation of this metabolite. However, formation of only the M-17 metabolite suggests that the isozyme present in the Caco-2 cells is distinct from CYP3A4, which also catalyzes the formation of significant quantities of the metabolites 9gamma-hydroxy cyclosporin A (M-1) and 4N-desmethyl cyclosporin A (M-21) from CsA. Interestingly, the amount of M-17 accumulating on the apical (AP) side was much greater than that on the basolateral (BL) side during the AP --> BL transport of CsA across Caco-2 cell monolayers. This is consistent with p-glycoprotein pump-mediated efflux of the metabolite to the apical side. Furthermore, formation of the M-17 metabolite on the AP side of cell monolayers during the AP --> BL transport of CsA was much greater than that during the BL --> AP transport. This result suggests that the p-glycoprotein efflux pump causes an increase in the metabolism of CsA during the course of its AP --> BL transport by effectively slowing down the transport of CsA molecules across Caco-2 cells. Thus, Caco-2 cells serve as an excellent model to dissect the relative roles played by p-glycoprotein-mediated efflux and CYP3A-catalyzed oxidation in modulating the overall absorption of CsA and other such compounds.

Journal Article
TL;DR: It is concluded that GSTA1-1 is the major isoform catalyzing busulfan conjugation, whereas GSTM-1 and GSTP-1 may be important in the protection of specific cells.
Abstract: Busulfan is eliminated by glutathione S-transferase (GST)-catalyzed conjugation with glutathione (GSH). We have characterized the busulfan-conjugating activity of purified human liver GSTA1-1, GSTA1-2, GSTA2-2, GSTM1-1, and placental GSTP1-1. Isoforms were purified from cytosol by GSH-affinity chromatography and chromatofocusing. In addition, the busulfan-conjugating activity of cDNA-expressed GTH1 and GTH2, corresponding to GSTA1-1 and GSTA2-2, were characterized. The major product of busulfan conjugation, a thiophenium ion (THT+), was assayed by GC/MS after conversion to tetrahydrothiophene (THT). THT+ formation rate increased linearly with busulfan concentration up to its solubility limit for all GST isoforms. Because Vmax and KM could not be determined separately, the slope of the velocity vs. substrate concentration plot, Vmax/KM was used to compare isoform activities. Vmax/KM for GSTA1-1 was 7.95 microliters/min/mg protein, the highest busulfan-conjugating activity of all human liver and placenta isoforms evaluated. GSTM1-1 and GSTP1-1, respectively, had 46% and 18% of the activity of GSTA1-1. Since the polymorphic mu-class GST catalyzed busulfan conjugation, we examined busulfan clearance in 50 patients undergoing high-dose busulfan before bone marrow transplantation. Busulfan clearance was normally distributed, suggesting that GSTM1-1 does not contribute significantly to the elimination of busulfan from the body. We conclude that GSTA1-1 is the major isoform catalyzing busulfan conjugation, whereas GSTM1-1 and GSTP1-1 may be important in the protection of specific cells.

Journal Article
TL;DR: Results from the chemical and immunochemical inhibition studies indicated the possible involvement of isoforms of the CYP3A subfamily in the oxidative metabolism of indinavir in rats, dogs, and monkeys, consistent with previous studies.
Abstract: Indinavir, a potent and specific inhibitor of human immunodeficiency virus protease, is undergoing clinical investigation for the treatment of acquired immunodeficiency syndrome. The studies described herein were designed to characterize the absorption, distribution, metabolism, and excretion of the drug in rats, dogs, and monkeys. Indinavir exhibited marked species differences in elimination kinetics. The plasma clearance was in the rank order: rat (107 ml/min/kg) > monkey (36 ml/min/kg) > dog (16 ml/min/kg). Significant differences in the bioavailability of indinavir also were observed. When given orally as a solution in 0.05 M citric acid, the bioavailability varied significantly from 72% in the dog to 19% in the monkey, and 24% in the rat. These differences in bioavailability were attributed mainly to species differences in the magnitude of hepatic first-pass metabolism. The distribution of indinavir was studied only in rats, both intravenously and orally. Intravenously, indinavir was distributed widely throughout the body. Brain uptake studies showed that indinavir penetrated the blood-brain barrier, but that the penetration was limited. After oral administration, indinavir was distributed rapidly into and out of the lymphatic system. The rapid lymph transfer is of clinical relevance, because a primary clinical hallmark of acquired immunodeficiency syndrome is the depletion of CD4 lymphocytes. Biliary and urinary recovery studies revealed that metabolism was the major route of indinavir elimination in all species, and N-dealkylation, N-oxidation, and hydroxylation seemed to be the major pathways. Although limited to qualitative aspects, the metabolite profile obtained from in vitro microsomal studies generally reflected the in vivo oxidative metabolism of indinavir in all species studies. Results from the chemical and immunochemical inhibition studies indicated the possible involvement of isoforms of the CYP3A subfamily in the oxidative metabolism of indinavir in rats, dogs, and monkeys. This is consistent with our previous studies, which have shown that CYP3A4 is the isoform responsible for the oxidative metabolism of indinavir in human liver microsomes. Furthermore, the in vivo oxidative metabolism of indinavir in rats, dogs, and monkeys was qualitatively similar to that in humans. The high degree of similarity in the metabolite profiles of drug metabolism between animals and humans validates the use of these animal models for toxicity studies of indinavir. Attempts were made to quantitatively extrapolate in vitro metabolic data to in vivo metabolism. With the application of the well-stirred and parallel-tube models, the hepatic clearance and hepatic extraction ratio were calculated using the in vitro Vmax/Km values. In rats, the predicted hepatic clearance (31 ml/ min/kg) and hepatic extraction ratio (0.47) agreed well with the observed in vivo hepatic clearance (43 ml/min/kg) and hepatic extraction ratio (0.68). In addition, the hepatic clearance of indinavir was predicted reasonably well in dogs and monkeys. Based on the in vitro intrinsic clearance of human liver microsomes, a small but significant hepatic first-pass metabolism (ca. 25%) is expected in humans.

Journal Article
TL;DR: Kinetic analysis showed that MK-639 is a very potent competitive inhibitor for testosterone 6beta-hydroxylation, with a Ki value of approximately 0.5 mu M, and results consistently indicate that CYP3A4 is the isoform responsible for the oxidative metabolism of MK- 639 in human liver microsomes.
Abstract: MK-639 (L-735,524) is a potent human immunodeficiency virus protease inhibitor under investigation in the treatment of acquired immunodeficiency syndrome. Five in vitro approaches have been used to identify the cytochrome P450 isoform(s) responsible for the human microsomal oxidative metabolism of MK-639. These approaches are: 1) chemical inhibition; 2) immunochemical inhibition; 3) metabolism by cDNA-expressed human cytochrome P450 enzymes; 4) a correlation analysis; and 5) competitive inhibition of marker activities. Ketoconazole and troleandomycin, both selective inhibitors for cytochrome P450 3A4 (CYP3A4), markedly inhibited the formation of all oxidative metabolites of MK-639; whereas other inhibitors (furafylline, sulfaphenazole, quinidine, S-mephenytoin, and diethyldithiocarbamate) had little effect on MK-639 metabolism. This suggested the involvement of CYP3A4 in MK-639 metabolism. Consistent with this, an anti-rat CYP3A1 rabbit polyclonal antibody, which shows a cross-reactive inhibition of CYP3A4-dependent testosterone 6beta-hydroxylation in human liver microsomes, completely inhibited MK-639 metabolism. Human recombinant CYP3A4 showed a high metabolic activity to form all MK-639 metabolites found in native human liver microsomes. In addition, the formation of individual MK-639 metabolites correlated well with each other and with testosterone 6beta-hydroxylation in 12 different human liver microsomes, whereas no correlation was observed between MK-639 metabolite formation and bufuralol 1'-hydroxylation (or tolbutamide methyl hydroxylation). Furthermore, MK-639 strongly inhibited testosterone 6beta-hydroxylation in a concentration-dependent manner. Kinetic analysis showed that MK-639 is a very potent competitive inhibitor for testosterone 6beta-hydroxylation, with a Ki value of approximately 0.5 mu M. Collectively, these results consistently indicate that CYP3A4 is the isoform responsible for the oxidative metabolism of MK-639 in human liver microsomes.

Journal Article
TL;DR: The common dietary constituent quercetin was a potent inhibitor of sulfoconjugation of acetaminophen and minoxidil by human liver cytosol, partially purified P- form phenolsulfotransferase (PST), and recombinant P-form PST, and a possible role for flavonoids as chemopreventive agents in sulfation-induced carcinogenesis is suggested.
Abstract: The common dietary constituent quercetin was a potent inhibitor of sulfoconjugation of acetaminophen and minoxidil by human liver cytosol, partially purified P-form phenolsulfotransferase (PST), and recombinant P-form PST, with IC50 values of 0.025-0.095 microM. Quercetin inhibition of acetaminophen was noncompetitive with respect to acceptor substrate, with a Ki value of 0.067 microM. A number of other flavonoids, such as fisetin, galangin, myricetin, kaempferol, chrysin, and apigenin, were also potent inhibitors of P-form PST-mediated sulfation, with IC50 values < 1 microM. Studies of structural analogs indicated the flavonoid 7-hydroxyl group as particularly important for potent inhibition. Potential human metabolites of quercetin were poor inhibitors. Curcumin, genistein, and ellagic acid (other polyphenolic natural products) were also inhibitors of P-form PST, with IC50 values of 0.38-34.8 microM. Quercetin was also shown to inhibit sulfoconjugation by the human hepatoma cell line Hep G2. Although less potent in this intact cell system (IC50 2-5 microM), quercetin was still more potent than 2,6-dichloro-4-nitrophenol, the classical P-form PST inhibitor that has been shown to be an inhibitor also in vivo. These observations suggest the potential for clinically important drug interactions, as well as a possible role for flavonoids as chemopreventive agents in sulfation-induced carcinogenesis.


Journal Article
TL;DR: Data suggest that CAI is processed into triazole and benzophenone moieties by phase I metabolism, and these metabolites or the parent compound may be conjugated for excretion by glucuronidation.
Abstract: The calcium influx inhibitor and cytostatic agent, 5-amino-1-1(4'-chlorobenzoyl-3,5-dichlorobenzyl)-1,2,3-triazole-4-carboxamide (CAI), is in phase I clinical trial for patients with refractory cancer. Additional chromatography peaks were observed during HPLC analysis of patient samples. Identification and characterization of physiological metabolites were undertaken using HPLC techniques developed for their purification from blood, pleural fluid, and urine samples. A hydrophobic metabolite, M1, was purified and functionally characterized. Structural analysis of the purified compound indicated that it is a 3,5-dichloro-4(p-chlorobenzoyl)-benzoic acid. Quantitative analysis of M1 concentration during CAI administration indicated that the rise in M1 concentration lagged behind that of CAI and persisted after CAI was no longer detectable. No clear relationship between CAI or M1 and either toxicity or efficacy was observed. Chromatography of patient blood and urine samples under conditions favoring hydrophilic metabolite detection suggested the presence of a glucuronide compound; this was also indicated by sample treatment with beta-glucuronidase. Attempts at purification did not yield a compound stable for structural analysis. The benzophenone metabolite, M1, was nonfunctional in assays of calcium influx inhibition or proliferation. No pharmacodynamic associations were observed for these metabolites, nor was there pharmacological activity of the M1 as an individual agent. These data suggest that CAI is processed into triazole and benzophenone moieties by phase I metabolism, and these metabolites or the parent compound may be conjugated for excretion by glucuronidation.

Journal Article
TL;DR: These studies indicate that the N-demethylation of S-mephenytoin by human liver microsomes is catalyzed primarily by CYP2B6.
Abstract: In vitro methods were used to identify the cytochrome P450 (CYP) enzyme(s) involved in S-mephenytoin N-demethylation. S-Mephenytoin (200 microM) was incubated with human liver microsomes, and nirvanol formation was quantitated by reversed-phase HPLC. S-Mephenytoin N-demethylase activity in a panel of human liver microsomes ranged 35-fold from 9 to 319 pmol/min/mg protein and correlated strongly with microsomal CYP2B6 activity (r = 0.91). Additional correlations were found with microsomal CYP2A6 and CYP3A4 activity (r = 0.88 and 0.74, respectively). Microsomes prepared from human beta-lymphoblastoid cells transformed with individual P450 cDNAs were assayed for S-mephenytoin N-demethylase activity. Of 11 P450 isoforms (P450s 1A1, 1A2, 2A6, 2B6, 2E1, 2D6, 2C8, 2C9, 2C19, 3A4, and 3A5) tested, only CYP2B6 catalyzed the N-demethylation of S-mephenytoin with an apparent K(m) of 564 microM. Experiments with P450 form-selective chemical inhibitors, competitive substrates, and anti-P450 antibodies were also performed. Troleandomycin, a mechanism-based CYP3A selective inhibitor, and coumarin, a substrate for CYP2A6 and therefore a potential competitive inhibitor, failed to inhibit human liver microsomal S-mephenytoin N-demethylation. In contrast, orphenadrine, an inhibitor of CYP2B forms, produced a 51 +/- 4% decrease in S-mephenytoin N-demethylase activity in human liver microsomes and a 45% decrease in recombinant microsomes expressing CYP2B6. Also, both CYP2B6-marker 7-ethoxytrifluoromethylcoumarin O-deethylase and S-mephenytoin N-demethylase activities were inhibited by approximately 65% by 5 mg anti-CYP2B1 IgG/mg microsomal protein. Finally, polyclonal antibody inhibitory to CYP3A1 failed to inhibit S-mephenytoin N-demethylase activity. Taken together, these studies indicate that the N-demethylation of S-mephenytoin by human liver microsomes is catalyzed primarily by CYP2B6.

Journal Article
TL;DR: Kinetic studies demonstrate that two forms of human liver cytochrome P450 are responsible for the formation of (R)-8-hydroxywarfarin and (S)-mephenytoin, and a high-affinity enzyme is identified as P4502C19 on the basis of the following evidence.
Abstract: Kinetic studies demonstrate that two forms of human liver cytochrome P450 are responsible for the formation of (R)-8-hydroxywarfarin: a low-affinity enzyme (KM approximately 1.5 mM), previously identified as P4501A2; and a high-affinity enzyme (KM = 330 microM), now identified as P4502C19 on the basis of the following evidence. In crossover inhibition studies with P4501A2-depleted human liver microsomes between (R)-warfarin and (S)-mephenytoin, reciprocal competitive inhibition was observed. Apparent KM values for (S)-mephenytoin-4'-hydroxylation (52-67 microM) were similar to the determined Ki values (58-62 microM) for (S)-mephenytoin inhibition of (R)-8-hydroxywarfarin formation. Similarly, the apparent KM for (R)-warfarin 8-hydroxylation in furafylline-pretreated microsomes (KM = 289-395 microM) was comparable with the Ki values (280-360 microM) for (R)-warfarin inhibition of (S)-4'-hydroxymephenytoin formation. Inhibition studies with tranylcypromine, a known inhibitor of (S)-mephenytoin hydroxylase activity, and either substrate in three different microsomal preparations yielded nearly identical inhibitory constants: Ki = 8.7 +/- 1.6 microM for inhibition of (S)-4'-hydroxymephenytoin formation and 8.8 +/- 2.5 microM for inhibition of (R)-8-hydroxywarfarin formation. In addition, fluconazole, a potent inhibitor of (R)-warfarin 8-hydroxylation, Ki = 2 microM, was found to inhibit (S)-mephenytoin hydroxylation with an identical Ki (2 microM). Finally, a strong correlation between (S)-mephenytoin 4-hydroxylation and (R)-warfarin 8-hydroxylation activities in furafylline-pretreated microsomes was demonstrated in 14 human liver microsomal preparations (r2 = 0.97).

Journal Article
TL;DR: These studies in hepatic microsomes demonstrate that fluoroquinolones can decrease CYP3A- and CYP1A-mediated biotransformation by competitive inhibition and that they have the potential to cause drug interactions with agents metabolized by these enzymes.
Abstract: The fluoroquinolone antibacterial agents have gained widespread use in the treatment of a broad range of bacterial infections. We recently described a possible interaction concerning the concomitant use of cyclosporine A and norfloxacin in pediatric renal transplant patients. We examined the effect of two common fluoroquinolone antibiotics on cytochrome P450-mediated drug biotransformations in human and rat liver microsomes. Rats were pretreated with inducers, which increased the levels of the P450 isozymes CYP3A2, CYP1A, CYP2E1, and CYP4A1. Ciprofloxacin and norfloxacin significantly depressed the N-demethylation of erythromycin by CYP3A4 in human microsomes and by CYP3A2 in rat microsomes. The inhibition was determined to be competitive in nature in rat microsomes, with ciprofloxacin and norfloxacin both exhibiting similar Ki values of 2.0 and 2.3 mM, respectively. Ciprofloxacin and norfloxacin also inhibited ethoxyresorufin-O-dealkylase (CYP1A). In contrast, ciprofloxacin and norfloxacin did not inhibit the metabolism of substrates that are specific for the P450 isozymes CYP2E1 and CYP4A1. Rats treated chronically with norfloxacin revealed no alterations in hepatic CYP3A2 protein levels or activity. These studies in hepatic microsomes demonstrate that fluoroquinolones can decrease CYP3A- and CYP1A-mediated biotransformation by competitive inhibition and that they have the potential to cause drug interactions with agents metabolized by these enzymes.

Journal Article
TL;DR: The results demonstrate the differential regulation of P 450s between the liver and small intestine, and provide a basis for further studies in assessing the potential of intestinal P450s to protect against orally ingested polycyclic aromatic hydrocarbon carcinogens.
Abstract: The composition and inducibility of cytochrome P450 (P450) in rat small intestinal epithelial cells were investigated with the use of RNA-polymerase chain reaction and immunoblot techniques. The complement of intestinal P450s is more restricted than hepatic forms. P450s 1A1, 2B1, and 3A1 were detected in enterocytes of untreated rats and were inducible by beta-naphthoflavone (BNF), phenobarbital, and pregnenolone-16alpha-carbonitrile or dexamethasone, respectively. In addition, P450s 2C6 and 2C11 were both constitutively expressed at low levels. In contrast, several P450 forms, which are found in the liver, were not detected in enterocytes of untreated or induced rats, including P450s 2A1, 2B2, 2E1, 3A2, and 4A1. P4501A2 mRNA was detected only in BNF-induced rat small intestine and at levels that did not result in its detectable translation. The most prominent inducible form in rat small intestine is P4501A1. Its inducibility diminishes markedly along the length of the small intestine from the duodenum to the ileum. Furthermore, the induction of P4501A1 in enterocytes was affected by the route of administration of the inducing agent. Thus, intestinal P4501A1 was more sensitive to orally administered BNF, whereas induction of hepatic P4501A1 was more sensitive to intraperitoneal administered BNF. Overall, the results demonstrate the differential regulation of P450s between the liver and small intestine, and provide a basis for further studies in assessing the potential of intestinal P450s to protect against orally ingested polycyclic aromatic hydrocarbon carcinogens.

Journal Article
TL;DR: Evidence is provided, in rats, for the generation of a reactive arene oxide species from CBZ, if not adequately detoxified, via conjugation with glutathione, this has the potential to initiate cellular damage.
Abstract: The metabolism of carbamazepine (CBZ) and its major metabolite in humans, carbamazepine 10,11-epoxide (CBZ-E), was examined in the rat in vivo. Particular emphasis was placed on the identification of dihydrohydroxythio adducts, which are detoxication products of reactive arene oxide intermediates. Anesthetized and cannulated male Wistar rats were administered [3H]CBZ (25 micrograms.kg-1 or 25 mg.kg-1) or [3H]CBZ-E (25 micrograms.kg-1 or 25 mg.kg-1) intravenously and bile and urine collected for 5 hr. Less than 8% of drug was excreted in the urine for each dosing regimen. Biliary excretion accounted for 73.7 +/- 6.2 and 41.8 +/- 6.2% (mean +/- SD, N = 4) of administered CBZ (25 micrograms.kg-1 and 25 mg.kg-1, respectively) and 47.6 (N = 2) and 28.1 +/- 6.0% of administered CBZ-E (25 micrograms.kg-1 and 25 mg.kg-1, respectively). The major route of metabolism of both CBZ and CBZ-E was N-glucuronidation. In rats given CBZ (25 mg.kg-1), the N-glucuronide of the parent compound accounted for 12.6 +/- 2.6% of the dose, whereas CBZ-E N-glucuronide accounted for 12.3 +/- 3.8% of the dose. At the lower dose of 25 micrograms.kg-1, these accounted for 18.6 +/- 3.0 and 36.7 +/- 4.7% of the dose, respectively. Similarly, for rats given CBZ-E (25 micrograms.kg-1), the N-glucuronide of the parent compound was the major metabolite, accounting for 19.1 +/- 4.5% of the dose. O-glucuronides were relatively minor metabolites of both drugs. Glutathione adducts were identified in the bile of both groups of animals. Although these adducts were relatively minor metabolites of CBZ-E (1.8% of the dose), they were more substantial products of the metabolism of CBZ. Three isometric glutathionyl dihydrohydroxy-CBZ adducts were identified by LC/MS. They collectively accounted for 5.8 +/- 0.9% of the dose. In conclusion, we have provided evidence, in rats, for the generation of a reactive arene oxide species from CBZ. If not adequately detoxified, via conjugation with glutathione, this has the potential to initiate cellular damage. In humans, a similar mechanism may be involved in CBZ-associated hypersensitivity.

Journal Article
TL;DR: The abundant expression of the CYP2A genes in rat and mouse olfactory tissue suggests a molecular basis for the known tissue-specific toxicity of numerous inhaled compounds in rodents.
Abstract: CYP2A10 and CYP2A11, which are abundant in olfactory microsomes from rabbits, are active in the metabolic activation of a number of nasal toxicants, such as hexamethylphosphoramide and N-nitrosodiethylamine. Previous immunohistochemical studies indicated that CYP2A-related cytochromes P450 may also be present in rodent and human olfactory tissue. In the present study, the expression of cytochromes P450 highly homologous to rabbit CYP2A10/11 in rat, mouse, and human nasal mucosa was studied. In Sprague-Dawley rats, CYP2A3 mRNA was detected in olfactory mucosa at levels much higher than those found in total RNA from lung. Similar observations were made for the level of microsomal CYP2A3 protein with the use of antibodies to rabbit CYP2A10/11. However, mRNAs for two other rat cytochrome P450 genes in the CYP2A subfamily, CYP2A1 and CYP2A2, were not detected in nasal tissue by RNA-polymerase chain reaction analysis. In C57BL/6 mice, both CYP2A4 and CYP2A5 mRNAs were detected in the olfactory mucosa by RNA-polymerase chain reaction, but the CYP2A5 transcript was present at a level much higher than that of CYP2A4. The expression of another mouse gene in CYP2A subfamily, CYP2A12, was not detected in nasal tissue. CYP2A5 protein was also detected in mouse olfactory microsomes at higher levels than in liver, lung, or kidney microsomes. However, no significant sex differences in the levels of CYP2A4/5 mRNA or microsomal coumarin 7-hydroxylase activity were found with the nasal tissue. In addition, consistent with previous immunohistochemical studies, the expression of CYP2A6 in human nasal mucosa was detected by RNA-polymerase chain reaction as well as RNA blot analysis. The identification of CYP2A6 in human nasal tissues may have important implications for risk assessment of potential nasal toxicants, and the abundant expression of the CYP2A genes in rat and mouse olfactory tissue suggests a molecular basis for the known tissue-specific toxicity of numerous inhaled compounds in rodents.

Journal Article
TL;DR: It is demonstrated that LPS administration induces P 4504A subfamily mRNA and P4504A3 protein expression and strain differences in both suppression and induction of P450s between the Sprague-Dawley and F344 rats are suggested.
Abstract: Bacterial lipopolysaccharide (LPS) has been previously shown to down-regulate the mRNA and protein expression of the hepatic cytochrome P450 (P450) isozymes 2C11 and 2C12. In this study, we examined the effects of LPS on the constitutive expression of P4503A2, P4502E1, and the P4504A subfamily in the rat. Fischer 344 and Sprague-Dawley rats were each administered 1 mg/kg LPS intraperitoneally and killed for hepatic RNA and microsome isolation at different times. LPS treatment was found to suppress P4502C11, P4503A2, and P4502E1 protein and mRNA expression in both strains of rat. Total microsomal P450 levels decreased by 30%, which was smaller than the effects on the levels of individual isozymes. The magnitude of suppression exhibited in the Sprague-Dawley rats, however, seemed to be more variable than that in the F344 strain. The mRNAs of all three of the P4504A subfamily members were induced 2- to 6-fold in the F344 rat livers after LPS administration. P4504A3 protein expression increased 2-fold, whereas P4504A1/2 protein levels decreased by 30%. Lauric acid omega-hydroxylase activity increased 1.6-fold in LPS-treated Fischer 344 rats and omega-1-hydroxylase activity decreased by 38%. In the Sprague-Dawley strain, however, decreases were seen in both omega- and omega-1-hydroxylase activities after LPS treatment. Our data demonstrate that LPS administration induces P4504A subfamily mRNA and P4504A3 protein expression. Furthermore, our findings also suggest strain differences in both suppression and induction of P450s between the Sprague-Dawley and F344 rats.

Journal Article
TL;DR: The present study provides the first direct confirmation, using human recombinant P450 enzymes and selective antibody inhibition, that CYP2C19 is a major high affinity OP 5-Hydroxylase and CYP3A4 is a low affinity OP-hydroxylating enzyme.
Abstract: Omeprazole (OP) is a potent antiulcer drug that is metabolized by liver cytochrome P450 (P450) enzymes. However, the identities of the P450 isoforms responsible for its metabolism have been controversial. 5-Hydroxyomeprazole (5OH-OP) formation cosegregates with the polymorphism of (S)-mephenytoin 4'-hydroxylation in humans, which is now known to be mediated by CYP2C19. Previous in vitro studies have indicated that liver microsomal 50H-OP formation correlates with both (S)-mephenytoin 4'-hydroxylase and CYP3A content. Inhibitor and CYP2C antibody studies also suggested that both enzymes may be involved in the 5-hydroxylation of OP, whereas CYP3A appears to be the predominant enzyme involved in OP sulfone (OP-S) formation. The present studies assessed the contribution of various CYP2C and CYP3A4 enzymes to OP metabolism by using recombinant human enzymes. CYP2C19, CYP2C8, CYP2C18, and CYP2C9 formed a single metabolite with an HPLC retention time identical to that of 5OH-OP. The turnover number for CYP2C19 was 13.4 +/- 1.4 nmol/min/nmol of P450, whereas those for CYP2C8, CYP2C18, and CYP2C9 were 2.2 +/- 0.1, 1.5 +/- 0.1, and approximately equal to 0.5 nmol/min/nmol of P450, respectively. Recombinant human CYP3A4 formed 5OH-OP and OP-S with turnover numbers of 5.7 +/- 1.1 and 7.4 +/- 0.9 nmol/min/nmol of P450, respectively, and formed a minor unidentified metabolite. CYP2C19 had a substantially lower KM for 5OH-OP formation than did CYP3A4, CYP2C8, or CYP2C18. Antibody to CYP2C proteins inhibited approximately equal to 70% of OP 5-hydroxylation at low substrate concentrations, comparable to those that may be encountered at therapeutically relevant doses, whereas antibody to CYP3A4 inhibited approximately equal to 30% of the activity. At high substrate concentrations, the contributions of the two enzymes to OP hydroxylation were roughly comparable (40-50%). In contrast, OP-S formation was completely inhibited by antibody to CYP3A4 proteins. The present study provides the first direct confirmation, using human recombinant P450 enzymes and selective antibody inhibition, that CYP2C19 is a major high affinity OP 5-hydroxylase and CYP3A4 is a low affinity OP-hydroxylating enzyme. The current work also shows, for the first time, that other CYP2C enzymes (CYP2C8, CYP2C9, and CYP2C18) may contribute to OP hydroxylation at high substrate concentrations. In contrast, OP-S was formed principally by CYP3A4.

Journal Article
TL;DR: Indinavir, a potent and specific inhibitor of the HIV-1(3 protease for the treatment of AIDS, was investigated in six healthy subjects after single oral administration of 400 mg and the metabolites in feces and urine suggested that the absorption of the drug was appreciable.
Abstract: Indinavir, N-[2(R)-hydroxy-1(S)-indanyl]-5-[2(S)-tertiary- butylaminocarbonyl-4-(3-pyridylmethyl)piperazino]-4(S)- hydroxy-2(R)-phenylmethylpentanamide (L-735,524,MK-639, ayl-4- Crixivan), is a potent and specific inhibitor of the HIV-1(3 protease for the treatment of AIDS. Disposition of [14C]indinavir was investigated in six healthy subjects after single oral administration of 400 mg. AUC, Cmax, and Tmax values for indinavir were 492 microM x min, 4.7 microM, and 50 min, respectively. The AUC value for the total radioactivity in plasma was 1.9 times higher than that of indinavir, indicating the presence of metabolites. The major excretory route was through feces, and the minor through urine. Mean recovery of radioactivity in the feces was 83.4%. In the urine, mean recoveries of the total radioactivity and unchanged indinavir were 18.7% and 11.0% of the dose, respectively. HPLC radioactivity and LC-MS/MS analyses of urine showed the presence of indinavir and low levels of quaternary pyridine N-glucuronide (M1), 2',3'-trans-dihydroxyindanylpyridine N-oxide (M2), 2',3'-trans-dihydroxyindan (M3) and pyridine N-oxide (M4a) analogs, and despyridylmethyl analogs of M3 (M5) and indinavir (M6). M5 and M6 were the major metabolites in urine. The metabolic profile in plasma was similar to that in urine. Quantitatively, the metabolites in feces accounted for >47% of the dose, which along with the urinary excretion of approximately 19%, suggested that the absorption of the drug was appreciable. In the feces, radioactivity was predominantly due to M3, M5, M6, and the parent compound. Thus, in urine and feces, the prominent metabolic pathways were oxidations and oxidative N-dealkylations. Excretion of the quaternary N-glucuronide metabolite in the urine, which is a minor metabolite in human, was specific to primates.

Journal Article
TL;DR: It is suggested that drugs are primarily metabolized by CYP2D6 will not exhibit racial differences in their disposition and there are no differences between Black subjects and White subjects in metabolism via CYP1D6.
Abstract: OBJECTIVE: The objective of this study was to determine whether metabolism via cytochrome P4502D6 (CYP2D6) was higher in Black subjects than White subjects. METHODS: Ten Black and 10 White healthy male volunteers who were phenotyped CYP2D6 extensive metabolizer phenotypes participated in this randomized, cross-over study in which metoprolol was used as a model CYP2D6 substrate. In both study phases, subjects received oral rac-metoprolol tartrate (200 mg); during one phase, subjects also took quinidine sulfate (100 mg) daily beginning 3 days before the dose of metoprolol. Plasma samples were collected for 12 and 24 hr after the dose in the metoprolol and metoprolol plus quinidine phases, respectively. Metoprolol enantiomer concentrations were determined by chiral HPLC with fluorescence detection. RESULTS: S-metoprolol areas under the concentration vs. time curves during the metoprolol phase were 879 +/- 600 ng/ml*hr in White subjects vs. 984 +/- 653 ng/ml*hr in Black subjects. During inhibition of CYP2D6-mediated metabolism with quinidine, S-metoprolol areas under the concentration vs. time curves were 2515 +/- 749 and 2719 +/- 742 in White and Black subjects, respectively. Metoprolol elimination half-lives in both groups were approximately doubled by quinidine. Mean S-metoprolol/R-metoprolol ratios were 1.39 in both racial groups during the metoprolol phase, and during the metoprolol plus quinidine phase were 0.89 and 1.03 in White subjects and Black subjects, respectively (p