Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Drug Metab Pharmacokinet ; 35(5): 432-440, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32807664

ABSTRACT

Uric acid is biosynthesized from purine by xanthine oxidase (XO) mainly in the liver and is excreted into urine and feces. Although several transporters responsible for renal and intestinal handling of uric acid have been reported, information on hepatic transporters is limited. In the present study, we studied quantitative contribution of transporters for hepatic handling of uric acid by mathematical modeling analysis in human sandwich-cultured hepatocytes (hSCH). Stable isotope-labeled hypoxanthine, hypoxanthine-13C2,15N (HX), was incubated with hSCH and formed 13C2,15N-labeled xanthine (XA) and uric acid (UA) were measured by LC-MS/MS time dependently. Rate constants for metabolism and efflux and uptake transport across sinusoidal and bile canalicular membranes of HX, XA and UA were estimated in the presence of inhibitors of XO and uric acid transporters. An XO inhibitor allopurinol significantly decreased metabolisms of HX and XA. Efflux into bile canalicular lumen was negligible and sinusoidal efflux was considered main efflux pathway of formed UA. Transporter inhibition study highlighted that GLUT9 strongly and MRP4 intermediately contribute to the sinusoidal efflux of UA with minor contribution of NPT1/4. Modeling analysis developed in the present study should be useful for quantitative prediction of uric acid disposition in liver.


Subject(s)
Hepatocytes/metabolism , Models, Biological , Uric Acid/metabolism , Cells, Cultured , Hepatocytes/cytology , Humans
2.
Drug Metab Dispos ; 47(3): 215-226, 2019 03.
Article in English | MEDLINE | ID: mdl-30593544

ABSTRACT

In the present study, the beagle dog was evaluated as a preclinical model to investigate organic anion transporting polypeptide (OATP)-mediated hepatic clearance. In vitro studies were performed with nine OATP substrates in three lots of plated male dog hepatocytes ± OATP inhibitor cocktail to determine total uptake clearance (CLuptake) and total and unbound cell-to-medium concentration ratio (Kpuu). In vivo intrinsic hepatic clearances (CLint,H) were determined following intravenous drug administration (0.1 mg/kg) in male beagle dogs. The in vitro parameters were compared with those previously reported in plated human, monkey, and rat hepatocytes; the ability of cross-species scaling factors to improve prediction of human in vivo clearance was assessed. CLuptake in dog hepatocytes ranged from 9.4 to 135 µl/min/106 cells for fexofenadine and telmisartan, respectively. Active process contributed >75% to CLuptake for 5/9 drugs. Rosuvastatin and valsartan showed Kpuu > 10, whereas cerivastatin, pitavastatin, repaglinide, and telmisartan had Kpuu < 5. The extent of hepatocellular binding in dog was consistent with other preclinical species and humans. The bias (2.73-fold) obtained from comparison of predicted versus in vivo dog CLint,H was applied as an average empirical scaling factor (ESFav) for in vitro-in vivo extrapolation of human CLint,H The ESFav based on dog reduced underprediction of human CLint,H for the same data set (geometric mean fold error = 2.1), highlighting its utility as a preclinical model to investigate OATP-mediated uptake. The ESFav from all preclinical species resulted in comparable improvement of human clearance prediction, in contrast to drug-specific empirical scalars, rationalized by species differences in expression and/or relative contribution of particular transporters to drug hepatic uptake.


Subject(s)
Drug Evaluation, Preclinical/methods , Metabolic Clearance Rate , Organic Anion Transporters/metabolism , Pharmaceutical Preparations/metabolism , Species Specificity , Animals , Dogs , Hepatocytes/metabolism , Humans , Infusions, Intravenous , Liver/cytology , Liver/metabolism , Male , Models, Animal , Models, Biological , Pharmaceutical Preparations/administration & dosage
3.
Drug Metab Dispos ; 46(5): 680-691, 2018 May.
Article in English | MEDLINE | ID: mdl-29352067

ABSTRACT

Functional interplay between transporters and drug-metabolizing enzymes is currently one of the hottest topics in the field of drug metabolism and pharmacokinetics. Uptake transporter-enzyme interplay is important to determine intrinsic hepatic clearance based on the extended clearance concept. Enzyme and efflux transporter interplay, which includes both sinusoidal (basolateral) and canalicular efflux transporters, determines the fate of metabolites formed in the liver. As sandwich-cultured hepatocytes (SCHs) maintain metabolic activities and form a canalicular network, the whole interplay between uptake and efflux transporters and drug-metabolizing enzymes can be investigated simultaneously. In this article, we review the utility and applicability of SCHs for mechanistic understanding of hepatic disposition of both parent drugs and metabolites. In addition, the utility of SCHs for mimicking species-specific disposition of parent drugs and metabolites in vivo is described. We also review application of SCHs for clinically relevant prediction of drug-drug interactions caused by drugs and metabolites. The usefulness of mathematical modeling of hepatic disposition of parent drugs and metabolites in SCHs is described to allow a quantitative understanding of an event in vitro and to develop a more advanced model to predict in vivo disposition.


Subject(s)
Hepatocytes/metabolism , Pharmaceutical Preparations/metabolism , Animals , Biological Transport/physiology , Cells, Cultured , Drug Interactions/physiology , Humans , Liver/metabolism , Membrane Transport Proteins/metabolism , Metabolic Clearance Rate/physiology
4.
J Med Chem ; 60(23): 9508-9530, 2017 12 14.
Article in English | MEDLINE | ID: mdl-29120624

ABSTRACT

The discovery of 1-({6-[(2-methoxy-4-propylbenzyl)oxy]-1-methyl-3,4-dihydronaphthalen-2-yl}methyl)azetidine-3-carboxylic acid 13n (ceralifimod, ONO-4641), a sphingosine-1-phosphate (S1P) receptor agonist selective for S1P1 and S1P5, is described. While it has been revealed that the modulation of the S1P1 receptor is an effective way to treat autoimmune diseases such as relapsing-remitting multiple sclerosis (RRMS), it was also reported that activation of the S1P3 receptor is implicated in some undesirable effects. We carried out a structure-activity relationship (SAR) study of hit compound 6 with an amino acid moiety in the hydrophilic head region. Following identification of a lead compound with a dihydronaphthalene central core by inducing conformational constraint, optimization of the lipophilic tail region led to the discovery of 13n as a clinical candidate that exhibited >30 000-fold selectivity for S1P1 over S1P3 and was potent in a peripheral lymphocyte lowering (PLL) test in mice (ED50 = 0.029 mg/kg, 24 h after oral dosing).


Subject(s)
Azetidines/pharmacology , Lymphocytes/drug effects , Naphthalenes/pharmacology , Receptors, Lysosphingolipid/agonists , Administration, Oral , Animals , Autoimmune Diseases/drug therapy , Azetidines/administration & dosage , Azetidines/chemistry , Azetidines/pharmacokinetics , CHO Cells , Cricetulus , Female , Humans , Macaca fascicularis , Male , Mice , Mice, Inbred BALB C , Naphthalenes/administration & dosage , Naphthalenes/chemistry , Naphthalenes/pharmacokinetics , Rats, Inbred Lew , Rats, Sprague-Dawley
5.
Drug Metab Dispos ; 44(1): 16-27, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26502773

ABSTRACT

To quantitatively understand the events in the human liver, we modeled a hepatic disposition of bosentan and its three known metabolites (Ro 48-5033, Ro 47-8634, and Ro 64-1056) in sandwich-cultured human hepatocytes based on the known metabolic pathway. In addition, the hepatotoxicity of Ro 47-8634 and Ro 64-1056 was investigated because bosentan is well known as a hepatotoxic drug. A model illustrating the hepatic disposition of bosentan and its three metabolites suggested the presence of a novel metabolic pathway(s) from the three metabolites. By performing in vitro metabolism studies on human liver microsomes, a novel metabolite (M4) was identified in Ro 47-8634 metabolism, and its structure was determined. Moreover, by incorporating the metabolic pathway of Ro 47-8634 to M4 into the model, the hepatic disposition of bosentan and its three metabolites was successfully estimated. In hepatocyte toxicity studies, the cell viability of human hepatocytes decreased after exposure to Ro 47-8634, and the observed hepatotoxicity was diminished by pretreatment with tienilic acid (CYP2C9-specific inactivator). Pretreatment with 1-aminobenzotriazole (broad cytochrome P450 inactivator) also tended to maintain the cell viability. Furthermore, Ro 64-1056 showed hepatotoxicity in a concentration-dependent manner. These results suggest that Ro 64-1056 is directly involved in bosentan-induced liver injury partly because CYP2C9 specifically mediates hydroxylation of the t-butyl group of Ro 47-8634. Our findings demonstrate the usefulness of a quantitative modeling of hepatic disposition of drugs and metabolites in sandwich-cultured hepatocytes. In addition, the newly identified metabolic pathway may be an alternative route that can avoid Ro 64-1056-induced liver injury.


Subject(s)
Chemical and Drug Induced Liver Injury/etiology , Endothelin Receptor Antagonists/metabolism , Endothelin Receptor Antagonists/toxicity , Hepatocytes/drug effects , Hepatocytes/enzymology , Models, Biological , Sulfonamides/metabolism , Sulfonamides/toxicity , Biological Transport , Biotransformation , Bosentan , Cell Culture Techniques , Cell Survival/drug effects , Cells, Cultured , Chemical and Drug Induced Liver Injury/enzymology , Chemical and Drug Induced Liver Injury/pathology , Cytochrome P-450 CYP2C9/metabolism , Cytochrome P-450 CYP2C9 Inhibitors/pharmacology , Female , Hepatocytes/pathology , Humans , Hydroxylation , Kinetics , Male , Microsomes, Liver/enzymology , Pyrimidines/metabolism , Pyrimidines/toxicity
6.
Drug Metab Pharmacokinet ; 30(2): 142-8, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25989889

ABSTRACT

A lower exposure of mycophenolic acid (MPA) in patients receiving MPA-mofetil in combination with cyclosporin A (CsA) is thought to be due to the inhibition of enterohepatic circulation of phenyl-glucuronide of MPA (MPAG). This study aimed to evaluate the interaction of CsA with hepatic disposition of MPA and MPAG in sandwich-cultured human hepatocytes (SCHH) by a mathematical modeling approach. In addition, the inhibition of CsA for glucuronidation of MPA to MPAG was examined in human liver microsomes. Inhibitory parameters of CsA for hepatic disposition of MPAG were estimated using a non-linear mixed effect model program, NONMEM. As a result, CsA did not influence the conversion of MPA to MPAG in either SCHH or human liver microsomes. In contrast, CsA inhibited the basolateral uptake of MPAG with an estimated maximum inhibitory effect (Imax) of 32.4%. CsA also inhibited basolateral efflux and biliary excretion of MPAG formed in SCHH, and the concentration producing 50% of Imax (IC50) for biliary excretion was lower than that for basolateral efflux. Our modeling approach suggests that CsA inhibits both basolateral uptake and biliary excretion of MPAG and leads to changes in systemic exposure of MPA and MPAG in humans.


Subject(s)
Cell Culture Techniques , Cyclosporine/pharmacology , Enterohepatic Circulation , Glucuronides/metabolism , Hepatocytes/metabolism , Membrane Transport Modulators/pharmacology , Membrane Transport Proteins/drug effects , Models, Biological , Mycophenolic Acid/analogs & derivatives , Biological Transport , Cells, Cultured , Dose-Response Relationship, Drug , Drug Interactions , Hepatobiliary Elimination/drug effects , Humans , Kinetics , Membrane Transport Proteins/metabolism , Microsomes, Liver/metabolism , Mycophenolic Acid/metabolism
7.
Mol Pharm ; 11(2): 568-79, 2014 Feb 03.
Article in English | MEDLINE | ID: mdl-24320552

ABSTRACT

In recent years, it has become increasingly important to test the safety of circulating metabolites of novel drugs as part of drug discovery and development programs. Accordingly, it is essential to develop suitable methods for identifying the major metabolites and their disposition in animal species and in humans. Mycophenolic acid (MPA), a selective inosine-5'-monophosphate dehydrogenase (IMPDH) inhibitor, is metabolized by glucuronidation and enterohepatic circulation of MPA-glucuronides is an important factor in the continuous systemic exposure of MPA. In humans, about 90% of the administered MPA dose is finally excreted as MPA phenyl-glucuronide (MPAG) in urine. Notably, the plasma concentration of MPAG is much higher than that of MPA. These factors suggest that, after its formation in hepatocytes, MPAG is excreted into bile and is also transported across the basolateral membrane to enter the circulation. In the present study, we performed metabolic/hepatobiliary transport studies of MPA and MPAG using sandwich-cultured human hepatocytes (SCHH) and constructed mathematical models of their hepatic disposition. We also performed vesicular transport studies to identify which human multidrug resistance-associated proteins (MRPs) are involved in the transport of MPAG from hepatocytes. MPAG was a preferred substrate for the biliary excretion transporter MRP2 and the hepatic basolateral transporters MRP3 and MRP4 in conventional and metabolic/hepatobiliary transport studies using SCHH and vesicular transport studies using human MRP-expressing membrane vesicles. The resulting mathematical model suggested that the basolateral transport plays an important role in the hepatic disposition of MPAG formed in hepatocytes. Our findings suggest that mathematical modeling of metabolic/hepatobiliary transport studies using SCH will provide useful information for determining the fate of metabolites formed in hepatocytes.


Subject(s)
Glucuronides/chemistry , Glucuronides/metabolism , Hepatocytes/metabolism , Models, Theoretical , Mycophenolic Acid/analogs & derivatives , Mycophenolic Acid/metabolism , Cells, Cultured , Humans , Liver/chemistry , Liver/metabolism , Mycophenolic Acid/chemistry
8.
Drug Metab Dispos ; 41(4): 735-43, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23303441

ABSTRACT

Paroxetine, a selective serotonin reuptake inhibitor, is metabolized in the liver and excreted into bile and urine as metabolites, but species differences have been observed in hepatic disposition between rats and humans. A major metabolite in rats is M1-glucuronide, whereas M1-glucuronide and M1-sulfate are found in humans. The primary excretion route of paroxetine-derived radioactivity in rats and humans is bile and urine, respectively. The aim of this study was to examine the usefulness of sandwich-cultured hepatocytes (SCH) to evaluate in vivo species differences of the hepatic disposition of paroxetine between rats and humans. The metabolite profile of [(3)H]paroxetine in SCH was similar to that in hepatocytes in suspension, and the in vitro metabolite profiles were similar to the published in vivo metabolic pathways for both species. Furthermore, the biliary excretion index (BEI) of formed M1-glucuronide in rat SCH (25.8-50.9%) was higher than that in human SCH (15.1-16.7%). The BEI of formed M1-sulfate (16.4-29.1%) was comparable to that of M1-glucuronide in human SCH, whereas the BEIs of paroxetine were negligible in SCH of both species. Moreover, M1-glucuronide was demonstrated to be a multidrug resistance-associated protein 2 substrate in both species, as determined by its uptake into ATP-binding cassette transporter-expressing membrane vesicles. SCH should prove to be useful to evaluate the processes of hepatic uptake and metabolism of parent drugs and the simultaneous examination of the biliary excretion of both parent drug and liver-derived metabolites.


Subject(s)
Cell Culture Techniques/methods , Hepatocytes/cytology , Hepatocytes/metabolism , Paroxetine/pharmacokinetics , Selective Serotonin Reuptake Inhibitors/pharmacokinetics , ATP-Binding Cassette Transporters/metabolism , Animals , Bile/metabolism , Humans , Male , Rats , Species Specificity , Tissue Distribution , Transport Vesicles/metabolism
9.
Toxicol Appl Pharmacol ; 263(2): 244-50, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22766462

ABSTRACT

We previously reported a quantitative time-lapse imaging (QTLI)-based analysis method to assess drug-drug interactions (DDI) at multidrug resistance-associated protein 2 (Mrp2) in rat sandwich-cultured hepatocyte (SCH) system, utilizing the fluorescent Mrp2 substrate, 5-(and 6)-carboxy-2',7'-dichlorofluorescein (CDF). Here, we aimed to examine the feasibility of using QTLI to evaluate DDI involving drug metabolite(s) generated in hepatocytes. We used estradiol (E2) and bilirubin as model compounds; both are not substrates of MRP2, whereas their hepatic metabolites, estradiol-17ß-glucuronide (E17G) or bilirubin glucuronides, are known to be its substrates as well as inhibitors. When rat SCHs were pre-exposed with E2, fluorescence of CDF accumulated in bile canaliculi decreased depending upon both the duration of pre-exposure and the concentration of extracellular E2. The decrease corresponded with the increase in intracellular concentration of E17G in hepatocytes. Furthermore, cytotoxicity of vinblastine, a substrate of MRP2, was enhanced in SCHs treated with E2. Similarly, CDF accumulated in bile canaliculi was significantly reduced in rat SCHs pre-exposed with bilirubin. In conclusion, these results suggest that phase II biotransformation of a competitor is reflected in alteration of MRP2-mediated CDF transport detected in QTLI. The QTLI might provide a convenient platform to evaluate transporter-based DDIs involving hepatic metabolites of drug candidates without the need to identify the metabolites.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Hepatocytes/metabolism , Time-Lapse Imaging/methods , Vinblastine/pharmacology , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Bilirubin/administration & dosage , Bilirubin/analogs & derivatives , Bilirubin/metabolism , Bilirubin/pharmacology , Cell Culture Techniques , Drug Interactions , Estradiol/administration & dosage , Estradiol/analogs & derivatives , Estradiol/metabolism , Estradiol/pharmacology , Feasibility Studies , Fluoresceins/chemistry , Male , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...