Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 114
Filter
Add more filters










Publication year range
1.
Xenobiotica ; 42(5): 483-95, 2012 May.
Article in English | MEDLINE | ID: mdl-22416982

ABSTRACT

It has been reported that hypertension exponentially increases in the patients with type 2 diabetes mellitus. Thus, this study was performed to investigate the pharmacokinetic and pharmacodynamic interactions between nifedipine and metformin, since both drugs were commonly metabolized via hepatic CYP2C and 3A subfamilies in rats. Nifedipine (3 mg/kg) and metformin (100 mg/kg) were simultaneously administered intravenously or orally to rats. Concentrations (I) of each drug in the liver and intestine, maximum velocity (V(max)), Michaelis-Menten constant (K(m)), and intrinsic clearance (CL(int)) for the disappearance of each drug, apparent inhibition constant (K(i)) and [I]/K(i) ratios of each drug in liver and intestine were determined. Also the metabolism of each drug in rat and human CYPs and blood pressure were also measured. After the simultaneous single intravenous administration of both drugs together, the AUCs of each drug were significantly greater than that in each drug alone due to the competitive inhibition for the metabolism of nifedipine by metformin via hepatic CYP3A1/2 and of metformin by nifedipine via hepatic CYP2C6 and 3A1/2. After the simultaneous single oral administration of both drugs, the significantly greater AUCs of each drug than that in each drug alone could have mainly been due to the competitive inhibition for the metabolism of nifedipine and metformin by each other via intestinal CYP3A1/2 in addition to competitive inhibition for the hepatic metabolism of each drug as same as the intravenous study.


Subject(s)
Cytochrome P-450 CYP3A/metabolism , Metformin/pharmacology , Metformin/pharmacokinetics , Nifedipine/pharmacology , Nifedipine/pharmacokinetics , Administration, Oral , Animals , Baculoviridae/drug effects , Baculoviridae/metabolism , Blood Proteins/metabolism , Cell Line , Dexamethasone/pharmacology , Drug Interactions , Humans , Injections, Intravenous , Intestinal Mucosa/metabolism , Intestines/drug effects , Isoenzymes/metabolism , Kinetics , Male , Metformin/administration & dosage , Metformin/metabolism , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Nifedipine/administration & dosage , Nifedipine/metabolism , Protein Binding/drug effects , Quinine/pharmacology , Rats , Rats, Sprague-Dawley , Sulfaphenazole/pharmacology , Troleandomycin/pharmacology
2.
Arch Pharm Res ; 34(10): 1729-34, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22076773

ABSTRACT

Docetaxel is a P-glycoprotein (P-gp) substrate and metabolized via cytochrome P450 (CYP) 3A subfamily in rats. Morin is an inhibitor of both CYPs and P-gp. Hence, the effects of morin on the intravenous and oral pharmacokinetics of docetaxel were investigated using 7,12-dimethylbenz[a]anthracene (DMBA)-induced mammary tumor rats (DMBA rats) as an animal model of human breast cancer. Docetaxel was administered intravenously (4 mg/kg) and orally (20 mg/kg) without and with morin (15 mg/kg) in DMBA rats. After the intravenous administration of docetaxel in control and DMBA rats with and without morin, the values of non-renal clearance and area under the plasma concentration-time (AUC) for docetaxel were comparable. Morin did not increase AUC or the absolute oral bioavailability (F) for docetaxel after the oral administration of docetaxel in control and DMBA rats with and without morin. The inhibition of hepatic and intestinal metabolism of docetaxel by morin and/or DMBA and the effect of intestinal P-gp inhibition by morin on the pharmacokinetics of docetaxel did not seem to be considerable in DMBA-induced mammary tumor rats.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacokinetics , Flavonoids/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Taxoids/pharmacokinetics , 9,10-Dimethyl-1,2-benzanthracene , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Administration, Oral , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Area Under Curve , Chromatography, High Pressure Liquid , Docetaxel , Female , Half-Life , Humans , Injections, Intravenous , Intestinal Mucosa/metabolism , Liver/metabolism , Mammary Neoplasms, Experimental/chemically induced , Mass Spectrometry , Rats , Taxoids/administration & dosage
3.
J Pharm Pharmacol ; 63(1): 49-57, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21155815

ABSTRACT

OBJECTIVES: Liver disease and acute renal failure (ARF) are closely associated. The pharmacokinetics of liquiritigenin (LQ), a candidate therapy for inflammatory liver disease, and its metabolites M1 and M2 were evaluated in rats with ARF induced by uranyl nitrate (U-ARF rats). METHODS: LQ was administered intravenously (20 mg/kg) or orally (50 mg/kg) in U-ARF and control rats, and uridine diphosphate-glucuronosyltransferases (UGT) activity and uridine 5'-diphosphoglucuronic acid (UDPGA) concentrations were determined in the liver and intestine. KEY FINDINGS: After intravenous LQ administration, U-ARF rats displayed significantly slower LQ renal clearance but no significant changes in the LQ area under the plasma concentration-time curve (AUC) compared with controls. This was because of similar hepatic UGT activity and UDPGA levels between two groups, which resulted in comparable non-renal clearance, as well as the limited contribution of LQ renal clearance to total LQ clearance. However, the AUC and AUC(M) /AUC(LQ) ratios of M1 and M2 were significantly increased in U-ARF rats because of decreased urinary excretion of M1 and M2. Similar results were observed following oral administration because of the comparable LQ intestinal metabolism in both groups and decreased urinary excretion of M1 and M2 in U-ARF rats. CONCLUSIONS: U-ARF rats displayed decreased urinary excretion of LQ glucuronides, resulting in significantly greater AUC and metabolite ratios of M1 and M2 following LQ administration.


Subject(s)
Acute Kidney Injury/complications , Flavanones/pharmacokinetics , Glucuronides/metabolism , Glucuronosyltransferase/metabolism , Administration, Oral , Animals , Area Under Curve , Flavanones/administration & dosage , Infusions, Intravenous , Male , Rats , Rats, Sprague-Dawley , Uranyl Nitrate/toxicity , Uridine Diphosphate Glucuronic Acid/metabolism
4.
Biopharm Drug Dispos ; 32(1): 38-49, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21162118

ABSTRACT

Hypertension is the most common comorbidity and major risk factor in patients with erectile dysfunction. The pharmacokinetics of mirodenafil, used for the treatment of erectile dysfunction, after the intravenous and oral administration (20 mg/kg) to 6-week-old rats (with blood pressure within the normotensive range) and 16-week-old spontaneously hypertensive rats (SHRs) and their age-matched control normotensive Kyoto-Wistar (KW) rats, and 16-week-old deoxycorticosterone acetate-salt-induced hypertensive rats (DOCA-salt rats) and their age-matched control Sprague-Dawley (SD) rats were compared. It was found that time-averaged renal clearance (Cl(r)) was of minor importance and that time-averaged non-renal clearance (Cl(nr)) was dominant. In both 6- and 16-week-old SHRs, the Cl(nr)s and areas under the curve (AUCs) of intravenous mirodenafil were significantly smaller and greater than those of the controls, but in 16-week-old DOCA-salt rats, they were comparable to the controls. Although the AUC of oral mirodenafil in 16-week-old SHRs was comparable to the controls, the Cl(nr)s (or total body clearances, Cls) of intravenous mirodenafil and intestinal intrinsic clearances were significantly smaller than the controls and comparable to the controls for both 6- and 16-week-old SHRs, unlike in the 16-week-old DOCA-salt rats. The above data suggest that the significantly smaller Cl(nr) and greater AUC of intravenous mirodenafil and comparable AUC of oral mirodenafil in 16-week-old SHR could be due to the hereditary characteristics of SHRs, and not due to the hypertensive state itself.


Subject(s)
Desoxycorticosterone/toxicity , Hypertension/metabolism , Pyrimidinones/pharmacokinetics , Sulfonamides/pharmacokinetics , Animals , Disease Models, Animal , Hypertension/chemically induced , Male , Pyrimidinones/metabolism , Random Allocation , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Rats, Sprague-Dawley , Sulfonamides/metabolism
5.
J Pharm Pharmacol ; 62(1): 1-23, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20722995

ABSTRACT

OBJECTIVES: In rats with diabetes mellitus induced by alloxan (DMIA) or streptozocin (DMIS), changes in the cytochrome P450 (CYP) isozymes in the liver, lung, kidney, intestine, brain, and testis have been reported based on Western blot analysis, Northern blot analysis, and various enzyme activities. Changes in phase II enzyme activities have been reported also. Hence, in this review, changes in the pharmacokinetics of drugs that were mainly conjugated and metabolized via CYPs or phase II isozymes in rats with DMIA or DMIS, as reported in various literature, have been explained. The changes in the pharmacokinetics of drugs that were mainly conjugated and mainly metabolized in the kidney, and that were excreted mainly via the kidney or bile in DMIA or DMIS rats were reviewed also. For drugs mainly metabolized via hepatic CYP isozymes, the changes in the total area under the plasma concentration-time curve from time zero to time infinity (AUC) of metabolites, AUC(metabolite)/AUC(parent drug) ratios, or the time-averaged nonrenal and total body clearances (CL(NR) and CL, respectively) of parent drugs as reported in the literature have been compared. KEY FINDINGS: After intravenous administration of drugs that were mainly metabolized via hepatic CYP isozymes, their hepatic clearances were found to be dependent on the in-vitro hepatic intrinsic clearance (CL(int)) for the disappearance of the parent drug (or in the formation of the metabolite), the free fractions of the drugs in the plasma, or the hepatic blood flow rate depending on their hepatic extraction ratios. The changes in the pharmacokinetics of drugs that were mainly conjugated and mainly metabolized via the kidney in DMIA or DMIS rats were dependent on the drugs. However, the biliary or renal CL values of drugs that were mainly excreted via the kidney or bile in DMIA or DMIS rats were faster. SUMMARY: Pharmacokinetic studies of drugs in patients with type I diabetes mellitus were scarce. Moreover, similar and different results for drug pharmacokinetics were obtained between diabetic rats and patients with type I diabetes mellitus. Thus, present experimental rat data should be extrapolated carefully in humans.


Subject(s)
Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 1/physiopathology , Pharmaceutical Preparations/metabolism , Alloxan , Animals , Area Under Curve , Cytochrome P-450 Enzyme System/metabolism , Humans , Rats , Species Specificity , Streptozocin
6.
J Pharm Pharmacol ; 62(8): 1084-8, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20663044

ABSTRACT

OBJECTIVES: It has been reported that docetaxel is a P-glycoprotein substrate and is metabolized via the cytochrome P450 (CYP) 3A subfamily in rats. Tesmilifene is a substrate of the CYP3A subfamily and is an inhibitor of P-glycoprotein. Thus, the effects of various doses of tesmilifene on the pharmacokinetics of intravenous and orally administered docetaxel have been investigated in rats. METHODS: Docetaxel (20 mg/kg as base) was administered intravenously and orally without and with tesmilifene (5, 10, and 20 mg/kg) in rats. KEY FINDINGS: After intravenous administration of docetaxel with tesmilifene, the values of nonrenal clearance (CL(NR)) and area under the plasma concentration-time (AUC) for docetaxel were comparable with those without tesmilifene. Tesmilifene did not increase the values of AUC or of absolute oral bioavailability (F) for docetaxel after oral administration of docetaxel with tesmilifene. CONCLUSIONS: The inhibition for the metabolism of docetaxel via hepatic and intestinal CYP3A subfamily, and inhibition of P-glycoprotein-mediated efflux of docetaxel in the intestine by tesmilifene were almost negligible. The extremely low value of F for docetaxel was due to the incomplete absorption from the gastrointestinal tract and considerable first-pass metabolism of docetaxel in rats.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Cytochrome P-450 CYP3A/metabolism , Phenyl Ethers/administration & dosage , Taxoids/administration & dosage , Taxoids/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Administration, Oral , Animals , Antineoplastic Agents/blood , Area Under Curve , Biotransformation , Docetaxel , Drug Interactions , Injections, Intravenous , Male , Metabolic Clearance Rate , Phenyl Ethers/metabolism , Rats , Rats, Sprague-Dawley , Taxoids/blood
7.
Biopharm Drug Dispos ; 31(5-6): 358-61, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20578211

ABSTRACT

The pharmacokinetics of SP-8203, a potential protective agent for the treatment of cerebral infarction, were evaluated after its intravenous (10, 20 and 30 mg/kg) and oral (10, 20, 30 and 100 mg/kg) administration in rats. After the intravenous administration of SP-8203, the AUCs of SP-8203 were dose-dependent; the dose-normalized AUCs were significantly greater with increasing doses. After the oral administration of SP-8203, plasma concentrations of SP-8203 were much lower than those after intravenous administration. This could be due to considerable hepatic and intestinal metabolism and the high percent of the dose recovered from the gastrointestinal tract (including its contents and feces) at 24 h as unchanged drug.


Subject(s)
Neuroprotective Agents/pharmacokinetics , Quinazolinones/administration & dosage , Quinazolinones/pharmacokinetics , Acetamides , Administration, Oral , Animals , Dose-Response Relationship, Drug , Infusions, Intravenous , Male , Neuroprotective Agents/administration & dosage , Rats , Rats, Sprague-Dawley
8.
Metabolism ; 59(10): 1472-80, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20170928

ABSTRACT

Liquiritigenin (LQ) is a candidate for the treatment of inflammatory liver disease. Many studies have confirmed that hepatic disease and diabetes mellitus are closely associated. Thus, the pharmacokinetic changes of LQ and its 2 glucuronides, M1 and M2, in a rat model of diabetes mellitus induced by streptozotocin (DMIS rats) were evaluated. Liquiritigenin was administered intravenously (20 mg/kg) or orally (50 mg/kg) in DMIS and control rats. Changes in in vitro activity and in vivo uridine 5'-diphosphoglucuronic acid level in the liver and intestine of DMIS rats compared with controls were also studied. After intravenous administration of LQ in DMIS rats, no significant changes in the pharmacokinetic parameters of LQ were observed. However, the AUC(M2)/AUC(LQ) ratio was significantly greater (by 53.0%) than that of controls. After oral administration of LQ, the AUC of LQ and metabolite ratios of M1 and M2 were comparable to controls. The increase in the formation of glucuronides of LQ, especially M2, after intravenous administration of LQ was due to the increased in vivo hepatic uridine 5'-diphosphoglucuronic acid level in DMIS rats as a result of alteration in carbohydrate metabolism in diabetes. The comparable pharmacokinetics of LQ, M1, and M2 after oral administration of LQ were mainly due to the comparable intestinal metabolism of LQ between the control and DMIS rats.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Flavanones/pharmacokinetics , Glucuronides/metabolism , Liver/metabolism , Uridine Diphosphate Glucuronic Acid/metabolism , Administration, Oral , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/pharmacokinetics , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/pathology , Drug Evaluation, Preclinical , Flavanones/administration & dosage , Flavanones/metabolism , Injections, Intravenous , Intestinal Mucosa/metabolism , Intestines/drug effects , Intestines/pathology , Liver/drug effects , Liver/pathology , Male , Microsomes, Liver/metabolism , Rats , Rats, Sprague-Dawley , Streptozocin
9.
Eur J Pharm Sci ; 39(1-3): 181-9, 2010 Jan 31.
Article in English | MEDLINE | ID: mdl-19961930

ABSTRACT

It has been reported that both liquiritigenin (LQ) and dimethyl-4,4'-dimethoxy-5,6,5',6'-dimethylenedioxybiphenyl-2,2'-dicarboxylate (DDB) have a hepatoprotective effect, and administration of both drugs together shows additive protective effect against acute liver injuries. Therefore, the pharmacokinetic interaction between LQ and DDB in rats was studied. LQ (20 and 50mg/kg for the i.v. and p.o. administration, respectively), DDB (10mg/kg for both i.v. and p.o. administration), and both drugs together were once administered intravenously or orally to rats. After the i.v. administration of both drugs together, the Cl(nr) and AUC of LQ were significantly faster (by 30.5%) and smaller (by 22.5%), respectively, than those of without DDB due to the faster hepatic blood flow rate by DDB. After the p.o. administration of both drugs together, the AUC of LQ was comparable to that of without DDB due to negligible effect of DDB on intestinal metabolism of LQ. The pharmacokinetic parameters of DDB after both i.v. and p.o. administration were not altered by LQ, indicating that LQ did not considerably affect the pharmacokinetics of DDB in rats.


Subject(s)
Biphenyl Compounds/pharmacokinetics , Dicarboxylic Acids/pharmacokinetics , Flavanones/pharmacokinetics , Liver/metabolism , Administration, Oral , Animals , Biphenyl Compounds/administration & dosage , Dicarboxylic Acids/administration & dosage , Drug Interactions , Flavanones/administration & dosage , Injections, Intravenous , Liver/blood supply , Liver/drug effects , Metabolic Clearance Rate , Rats , Rats, Sprague-Dawley , Regional Blood Flow
10.
Eur J Pharm Sci ; 39(1-3): 1-7, 2010 Jan 31.
Article in English | MEDLINE | ID: mdl-19454315

ABSTRACT

It has been reported that metformin was primarily metabolized via hepatic CYP2C11, 2D1, and 3A1/2 in rats. It has also been reported that the protein expression and/or mRNA levels of hepatic CYP2C11, 2D subfamily, and 3A1 have decreased, decreased, and increased, respectively, in U-ARF rats. Thus, pharmacokinetic changes of intravenous metformin in U-ARF rats were evaluated. Metformin was administered intravenously at a dose of 50mg/kg to control and U-ARF rats. After i.v. administration of metformin to U-ARF rats, its time-averaged total body clearance was significantly slower (95.2% decrease) than controls. This could have been due to both significantly slower time-averaged renal clearance (99.1% decrease; due to a urine flow rate-dependent timed-interval renal clearance of the drug, a decrease in renal OCT2, and/or an impaired kidney function in U-ARF rats) and time-averaged non-renal clearance (83.8% decrease; due to a decrease in hepatic CYP2C11 and 2D subfamily in U-ARF rats).


Subject(s)
Acute Kidney Injury/metabolism , Metformin/pharmacokinetics , Acute Kidney Injury/chemically induced , Animals , Disease Models, Animal , Injections, Intravenous , Kidney Function Tests , Liver Function Tests , Male , Metabolic Clearance Rate , Metformin/administration & dosage , Microsomes, Liver/metabolism , Rats , Rats, Sprague-Dawley , Uranyl Nitrate
11.
J Pharm Pharmacol ; 61(12): 1637-42, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19958586

ABSTRACT

OBJECTIVES: It has been reported that hepatic cytochrome P450 (CYP)2C9 and CYP3A4 are responsible for the metabolism of sildenafil and formation of its metabolite, N-desmethylsildenafil, in humans. However, in-vivo studies in rats have not been reported. METHODS: Sildenafil (20 mg/kg) was administered intravenously to rats pretreated with sulfaphenazole, cimetidine, quinine hydrochloride or troleandomycin, inhibitors of CYP2C6, CYP2C11, CYP2D subfamily and CYP3A1/2, respectively. In-vitro studies using rat liver microsomes were also performed. KEY FINDINGS: The area under the plasma-concentration time curve (AUC) was increased and clearance of sildenafil decreased in rats pretreated with cimetidine or troleandomycin. The AUC ratio for N-desmethylsildenafil (0-4 h) : sildenafil (0-infinity) was significantly decreased only in rats pretreated with cimetidine. Similar results were obtained in the in-vitro study using rat liver microsomes. CONCLUSIONS: Sildenafil is metabolised via hepatic CYP2C11 and 3A1/2, and N-desmethylsildenafil is mainly formed via hepatic CYP2C11 in rats. Thus, rats could be a good model for pharmacokinetic studies of sildenafil and N-desmethylsildenafil in humans.


Subject(s)
Cimetidine/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Enzyme Inhibitors/pharmacology , Liver/metabolism , Phosphodiesterase Inhibitors/pharmacokinetics , Piperazines/pharmacokinetics , Sulfones/pharmacokinetics , Troleandomycin/pharmacology , Animals , Area Under Curve , Inactivation, Metabolic , Male , Microsomes , Phosphodiesterase Inhibitors/metabolism , Piperazines/metabolism , Purines/metabolism , Purines/pharmacokinetics , Rats , Rats, Sprague-Dawley , Sildenafil Citrate , Sulfones/metabolism
12.
J Pharm Pharmacol ; 61(10): 1325-32, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19814864

ABSTRACT

OBJECTIVES: It has been reported that mirodenafil is primarily metabolized via hepatic cytochrome P450 (CYP) 1A1/2, 2B1/2, 2D1 and 3A1/2 in rats. It has also been reported that the protein expression of hepatic CYP3A1 and intestinal CYP1A1 and 3A1/2 increases and that of hepatic CYP2D1 decreases in rats with acute renal failure induced by uranyl nitrate (U-ARF rats). Thus, the pharmacokinetics of mirodenafil were studied in control and U-ARF rats. METHODS: The pharmacokinetic parameters of mirodenafil and SK3541 (a metabolite of mirodenafil) were compared after the intravenous and oral administration of mirodenafil at a dose of 20 mg/kg to U-ARF and control rats. KEY FINDINGS: After interavenous administration of mirodenafil to U-ARF rats, the total area under the concentration-time curve (AUC) of mirodenafil was significantly smaller (36.5% decrease) than controls, possibly due to the significantly faster non-renal clearance (66.1% increase; because of increase in the protein expression of hepatic CYP3A1) than controls. After the oral administration of mirodenafil to U-ARF rats, the AUC of mirodenafil was also significantly smaller (47.8% decrease) due to the increase in the protein expression of hepatic CYP3A1 and intestinal CYP1A1 and 3A1/2 compared with controls. CONCLUSIONS: After both intravenous and oral administration of mirodenafil to U-ARF rats, the AUC(SK3541)/AUC(mirodenafil) ratios were comparable with that in controls and this could be due to further metabolism of SK3541 in rats.


Subject(s)
Acute Kidney Injury/metabolism , Aryl Hydrocarbon Hydroxylases/metabolism , Cytochrome P-450 CYP1A1/metabolism , Liver/metabolism , Pyrimidinones/pharmacokinetics , Sulfonamides/pharmacokinetics , Acute Kidney Injury/chemically induced , Administration, Oral , Animals , Cytochrome P-450 CYP3A , Disease Models, Animal , Injections, Intravenous , Intestinal Mucosa/metabolism , Male , Microsomes/metabolism , Pyrimidinones/administration & dosage , Rats , Rats, Sprague-Dawley , Sulfonamides/administration & dosage , Uranyl Nitrate
13.
Eur J Pharm Sci ; 38(5): 465-71, 2009 Dec 08.
Article in English | MEDLINE | ID: mdl-19761842

ABSTRACT

Ipriflavone was reported to be primarily metabolized via hepatic cytochrome P450 (CYP) 1A1/2 and 2C11 in male Sprague-Dawley rats. The protein expression and/or mRNA levels of hepatic CYP1A subfamily and 2C11 was reported to be increased and decreased, respectively, in diabetic rats induced by streptozotocin (DMIS rats). Thus, the pharmacokinetic parameters of ipriflavone and its two metabolites, M1 and M5, were compared after the i.v. (20mg/kg) and p.o. (200mg/kg) administration of ipriflavone to control and DMIS rats. After both i.v. and p.o. administration of ipriflavone to DMIS rats, the AUCs of ipriflavone were significantly smaller (by 31.7% and 34.2% for i.v. and p.o. administration, respectively) than controls. The faster Cl(nr) (smaller AUC) of i.v. ipriflavone could have been due to the faster hepatic Cl(int) (because of an increase in the protein expression and/or mRNA level of hepatic CYP1A subfamily) and the faster hepatic blood flow rate than controls. The smaller AUC of p.o. ipriflavone in DMIS rats could have mainly been due to the faster intestinal Cl(int) (because of an increase in the intestinal CYP1A subfamily) than controls.


Subject(s)
Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/drug therapy , Disease Models, Animal , Isoflavones/administration & dosage , Isoflavones/pharmacokinetics , Streptozocin/toxicity , Administration, Oral , Animals , Injections, Intravenous , Isoflavones/metabolism , Male , Metabolic Clearance Rate/drug effects , Metabolic Clearance Rate/physiology , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Rats , Rats, Sprague-Dawley
14.
Biopharm Drug Dispos ; 30(8): 485-93, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19753555

ABSTRACT

It has been reported that chlorzoxazone (CZX) was primarily metabolized via hepatic Cyp2e1 to form 6-hydroxychlorzoxazone (OH-CZX) in rats, and the activity of aniline hydroxylase (a Cyp2e1 marker) in the liver was significantly decreased in rats at 24 h after pretreatment with lipopolysaccharide derived from Klebsiella pneumoniae (24 h KPLPS rats), whereas the levels were not changed at 2 h and 96 h in the KPLPS rats. Thus, the time-dependent pharmacokinetic parameters of CZX and OH-CZX were evaluated after the intravenous administration of CZX (20 mg/kg) to control rats, and the 2 h, 24 h and 96 h KPLPS rats along with the time-dependent changes in the protein expression of hepatic Cyp2e1. After the intravenous administration of CZX to 24 h KPLPS rats, the AUC(0-2 h) of OH-CZX and AUC(OH-CZX, 0-2 h)/AUC(CZX) were significantly smaller (by 40.5% and 71.2%, respectively) than those of controls due to the significant decrease (by 75.3%) in the protein expression of hepatic Cyp2e1. However, in 96 h KPLPS rats, the pharmacokinetic parameters of both CZX and OH-CZX were unchanged compared with controls due to the restoration of the protein expression of hepatic Cyp2e1 to control levels. These observations highlighted the existence of the time-dependent effects of KPLPS on the pharmacokinetics of CZX and OH-CZX in rats.


Subject(s)
Chlorzoxazone/analogs & derivatives , Cytochrome P-450 CYP2E1/metabolism , Endotoxins/pharmacology , Klebsiella pneumoniae/chemistry , Lipopolysaccharides/pharmacology , Acute Kidney Injury/metabolism , Animals , Chlorzoxazone/pharmacokinetics , Disease Models, Animal , Drug Interactions , Injections, Intravenous , Lipopolysaccharides/pharmacokinetics , Male , Microsomes, Liver/metabolism , Muscle Relaxants, Central/pharmacokinetics , Rats , Rats, Sprague-Dawley
15.
Biopharm Drug Dispos ; 30(6): 305-17, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19639551

ABSTRACT

The pharmacokinetics of mirodenafil and its two metabolites, SK3541 and SK3544, after intravenous (5, 10, 20 and 50 mg/kg) and oral (10, 20 and 50 mg/kg) administration of mirodenafil, and the first-pass effect of mirodenafil after intravenous, oral, intraportal, intragastric and intraduodenal (20 mg/kg) administration of mirodenafil were evaluated in rats. The pharmacokinetics of mirodenafil and SK3541 were dose-dependent after both intravenous and oral administration of mirodenafil due to the saturable hepatic metabolism of mirodenafil. After oral administration of mirodenafil, approximately 2.59% of the oral dose was not absorbed, the F value was approximately 29.4%, and the hepatic and gastrointestinal first-pass effects of mirodenafil were approximately 21.4% and 54.3% of the oral dose, respectively. The low F value of mirodenafil in rats was mainly due to considerable hepatic and gastrointestinal first-pass effects in rats. The equilibrium plasma-to-blood cell partition ratios of mirodenafil were independent of the initial blood mirodenafil concentrations of 1-10 microg/ml; the mean values were 1.08-1.21. The plasma binding values of mirodenafil to rat plasma was 87.8%.


Subject(s)
Penile Erection/physiology , Phosphodiesterase 5 Inhibitors , Pyrimidinones/administration & dosage , Pyrimidinones/pharmacokinetics , Sulfonamides/administration & dosage , Sulfonamides/pharmacokinetics , Animals , Dose-Response Relationship, Drug , Male , Penile Erection/drug effects , Pyrimidinones/chemistry , Rats , Rats, Sprague-Dawley , Sulfonamides/chemistry , Tissue Distribution/drug effects , Tissue Distribution/physiology
16.
Biopharm Drug Dispos ; 30(6): 294-304, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19591122

ABSTRACT

Ipriflavone, a derivative of naturally occurring isoflavones, was primarily metabolized in rats via hepatic CYP1A1/2 and 2C11. Protein and mRNA expression of CYP1A2 in the liver, reported to be increased in mutant Nagase analbuminemic rats (NARs), should influence the pharmacokinetic parameters of ipriflavone. In this study, the contribution of hepatic CYP2C11 and intestinal CYP1A protein to the metabolism and the pharmacokinetic parameters of ipriflavone were examined after intravenous (20 mg/kg) and oral (200 mg/kg) administration to male Sprague-Dawley (control) rats and NARs. There was no change in the protein expression of hepatic CYP2C11. By contrast, CYP1A protein of the intestine increased by almost 100%. After the intravenous administration of ipriflavone to NARs, the Cl(nr) and AUC were unchanged, suggesting that the contribution of the increase in protein expression and mRNA level of hepatic CYP1A2 to hepatic metabolism of the drug in NARs seemed to be almost negligible. However, after the oral administration of ipriflavone to NARs, the AUC was significantly lower than that in the control rats (53.0% decrease), possibly due to the increased intestinal CYP1A that resulted in increased intestinal metabolism and decreased gastrointestinal absorption of ipriflavone in NARs.


Subject(s)
Acetylglucosaminidase/genetics , Acetylglucosaminidase/metabolism , Isoflavones/pharmacokinetics , Mutation/physiology , Animals , Aryl Hydrocarbon Hydroxylases/genetics , Aryl Hydrocarbon Hydroxylases/metabolism , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P450 Family 2 , Male , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Rats , Rats, Mutant Strains , Rats, Sprague-Dawley , Steroid 16-alpha-Hydroxylase/genetics , Steroid 16-alpha-Hydroxylase/metabolism
17.
Biopharm Drug Dispos ; 30(3): 107-16, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19288444

ABSTRACT

It is well known that there are various changes in the expression of hepatic and intestinal CYPs in mutant Nagase analbuminemic rats (NARs). It has been reported that the protein expression of hepatic CYP1A2 was increased, whereas that of hepatic CYP3A1 was not altered, and it was also found that the protein expression of the intestinal CYP1A subfamily significantly increased in NARs from our other study. In addition, in this study additional information about CYP changes in NARs was obtained; the protein expression of the hepatic CYP2D subfamily was not altered, but that of the intestinal CYP3A subfamily increased in NARs. Because omeprazole is metabolized via hepatic CYP1A1/2, 2D1, 3A1/2 in rats, it could be expected that the pharmacokinetics of omeprazole would be altered in NARs. After intravenous administration of omeprazole to NARs, the Cl(nr) was significantly faster than in the controls (110 versus 46.6 ml/min/kg), and this could be due to an increase in hepatic metabolism caused by a greater hepatic CYP1A2 level in addition to greater free fractions of the drug in NARs. After oral administration of omeprazole to NARs, the AUC was also significantly smaller (80.1% decrease) and F was decreased in NARs. This could be primarily due to increased hepatic and intestinal metabolism caused by greater hepatic CYP1A2 and intestinal CYP1A and 3A levels. In particular, the smaller F could mainly result from greater hepatic and intestinal first-pass effect in NARs than in the controls.


Subject(s)
Acetylglucosaminidase/metabolism , Cytochrome P-450 CYP1A2/metabolism , Liver/enzymology , Omeprazole/pharmacokinetics , Proton Pump Inhibitors/pharmacokinetics , Acetylglucosaminidase/genetics , Administration, Oral , Animals , Area Under Curve , Cytochrome P-450 CYP3A/metabolism , Cytochromes , Infusions, Intravenous , Intestines/enzymology , Isoenzymes , Male , Microsomes, Liver/enzymology , Mutation , Omeprazole/administration & dosage , Protein Binding , Proton Pump Inhibitors/administration & dosage , Rats , Rats, Mutant Strains , Rats, Sprague-Dawley , Up-Regulation
18.
J Pharm Sci ; 98(11): 4327-42, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19226634

ABSTRACT

Pharmacokinetics of liquiritigenin (LQ) and its two glucuronide metabolites, M1 and M2, in mice, rats, rabbits, and dogs and animal scale-up of the pharmacokinetic parameters of LQ were evaluated. After intravenous administration of LQ, the AUC (AUC(0-t)) values of LQ, M1, and M2 were proportional to LQ doses in all animals studied. Animal scale-up of some pharmacokinetic parameters of LQ was performed based on the parameters after its intravenous administration (20 mg/kg; in the linear pharmacokinetic range) to the four species. Linear relationships were obtained (r > 0.968) between log CL (or CL/f(u)) (L/h) and log species body weight (W) (kg) [CL (or CL/f(u)) = 3.29 (34.0) W(0.723 (0.789))] and log V(ss) (or V(ss)/f(u)) (L) and log W (kg) [V(ss) (or V(ss)/f(u)) = 0.340 (3.52) W(0.882 (0.948))]. Interspecies scale-up of plasma concentration-time data of LQ using apolysichron (complex Dedrick plots) resulted in similar profiles, and plasma concentration-time profile of humans were predicted using the well-fitted four animal data. Our results indicate that the LQ data obtained from laboratory animals could be utilized to generate preliminary estimates of the pharmacokinetic parameters of LQ in humans. These parameters can serve as guidelines for better planning of clinical studies.


Subject(s)
Flavanones/pharmacokinetics , Administration, Oral , Animals , Area Under Curve , Biological Availability , Body Weight , Dogs , Dose-Response Relationship, Drug , Flavanones/administration & dosage , Flavanones/blood , Half-Life , Humans , Infusions, Intravenous , Male , Metabolic Clearance Rate , Mice , Mice, Inbred ICR , Predictive Value of Tests , Protein Binding , Rabbits , Rats , Rats, Sprague-Dawley , Species Specificity
19.
J Pharm Pharmacol ; 61(1): 47-54, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19126296

ABSTRACT

OBJECTIVES: It has been reported that the non-renal clearance of furosemide was significantly faster in rats pretreated with phenobarbital but was not altered in rats pretreated with 3-methylcholanthrene. However, no studies on other cytochrome P450 (CYP) isozymes have yet been reported in rats. METHOD: Furosemide 20 mg/kg was administered intravenously to rats pretreated with various CYP inducers--3-methylcholanthrene, orphenadrine citrate and isoniazid, inducers of CYP1A1/2, 2B1/2 and 2E1, respectively, in rats--and inhibitors--SKF-525A (a non-specific inhibitor of CYP isozymes), sulfaphenazole, cimetidine, quinine hydrochloride and troleandomycin, inhibitors of CYP2C6, 2C11, 2D and 3A1/2, respectively, in rats. KEY FINDINGS: The non-renal clearance of furosemide was significantly faster (55.9% increase) in rats pretreated with isoniazid, but slower in those pretreated with cimetidine or troleandomycin (38.5% and 22.7% decreases, respectively), than controls. After incubation of furosemide with baculovirus-infected insect cells expressing CYP2C11, 2E1, 3A1 or 3A2, furosemide was metabolized via CYP2C11, 2E1, 3A1 and 3A2. CONCLUSIONS: These findings could help explain possible pharmacokinetic changes of furosemide in various rat disease models (where CYP2C11, 2E1, 3A1 and/or CYP3A2 are altered) and drug-drug interactions between furosemide and other drugs (mainly metabolized via CYP2C11, 2E1, 3A1 and/or 3A2).


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Enzyme Activators/pharmacology , Enzyme Inhibitors/pharmacology , Furosemide/pharmacokinetics , Animals , Area Under Curve , Aryl Hydrocarbon Hydroxylases/metabolism , Cimetidine/administration & dosage , Cimetidine/pharmacology , Diuretics/administration & dosage , Diuretics/metabolism , Diuretics/pharmacokinetics , Drug Interactions , Enzyme Activators/administration & dosage , Enzyme Inhibitors/administration & dosage , Furosemide/administration & dosage , Furosemide/metabolism , Half-Life , Infusions, Intravenous , Injections, Intravenous , Isoniazid/administration & dosage , Isoniazid/pharmacology , Male , Methylcholanthrene/administration & dosage , Methylcholanthrene/pharmacology , Orphenadrine/administration & dosage , Orphenadrine/pharmacology , Proadifen/administration & dosage , Proadifen/pharmacokinetics , Quinine/administration & dosage , Quinine/pharmacology , Rats , Rats, Sprague-Dawley , Troleandomycin/administration & dosage , Troleandomycin/pharmacology , Weight Gain/drug effects
20.
J Pharm Sci ; 98(4): 1587-601, 2009 Apr.
Article in English | MEDLINE | ID: mdl-18780336

ABSTRACT

Pharmacokinetics of amitriptyline and nortriptyline were evaluated after intravenous (2.5-10 mg/kg) and oral (10-100 mg/kg) administration of amitriptyline to rats. The hepatic, gastric, and intestinal first-pass effects of amitriptyline were also measured at a dose of 10 mg/kg. The areas under the plasma concentration-time curve (AUCs) of amitriptyline were dose-proportional following both intravenous and oral administration. After oral administration of amitriptyline, approximately 1.50% of the dose was not absorbed, the extent of absolute oral bioavalability (F) was approximately 6.30%, and the hepatic and intestinal first-pass effects of amitriptyline were approximately 9% and 87% of the oral dose, respectively. Although the hepatic first-pass effect was 78.9% after absorption into the portal vein, the value was only 9% of the oral dose due to considerable intestinal first-pass effect in rats. The low F of amitriptyline in rats was primarily attributable to considerable intestinal first-pass effect. This study proves the little contribution of considerable hepatic first-pass effect to low F of amitriptyline due to great intestinal first-pass effect in rats. The lower F value of amitriptyline in rats than that in humans (46 +/- 48%) was due to grater metabolism of amitriptyline in rats' liver and/or small intestine.


Subject(s)
Amitriptyline/pharmacokinetics , Intestine, Small/metabolism , Liver/metabolism , Nortriptyline/pharmacokinetics , Administration, Oral , Amitriptyline/administration & dosage , Amitriptyline/blood , Animals , Biological Availability , Dose-Response Relationship, Drug , Gastric Mucosa/metabolism , Injections, Intravenous , Male , Nortriptyline/administration & dosage , Nortriptyline/blood , Rats , Rats, Sprague-Dawley , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...