Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters










Publication year range
1.
Xenobiotica ; 44(7): 635-43, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24417753

ABSTRACT

1.Few studies describing the pharmacokinetic properties of chlorogenic acid (CA) and corydaline (CRD) which are marker compounds of a new prokinetic botanical agent, DA-9701, have been reported. The aim of the present study is to evaluate the pharmacokinetic properties CA and CRD following intravenous and oral administration of pure CA (1-8 mg/kg) or CRD (1.1-4.5 mg/kg) and their equivalent dose of DA-9701 to rats. 2. Dose-proportional AUC and dose-independent clearance (10.3-12.1 ml/min/kg) of CA were observed following its administration. Oral administration of CA as DA-9701 did not influence the oral pharmacokinetic parameters of CA. Incomplete absorption of CA, its decomposition in the gastrointestinal tract, and/or pre-systemic metabolism resulted in extremely low oral bioavailability (F) of CA (0.478-0.899%). 3. CRD showed greater dose-normalized AUC in the higher dose group than that in lower dose group(s) after its administration due to saturation of its metabolism via decreased non-renal clearance (by 51.3%) and first-pass extraction. As a result, the F of CRD following 4.5 mg/kg oral CRD (21.1%) was considerably greater than those of the lower dose groups (9.10 and 13.8%). However, oral administration of CRD as DA-9701 showed linear pharmacokinetics as a result of increased AUC and F in lower-dose groups (by 182% and 78.5%, respectively) compared to those of pure CRD. The greater oral AUC of CRD for DA-9701 than for pure CRD could be due to decreased hepatic and/or GI first-pass extraction of CRD by other components in DA-9701.


Subject(s)
Berberine Alkaloids/pharmacokinetics , Chlorogenic Acid/pharmacokinetics , Plant Preparations/pharmacokinetics , Administration, Oral , Animals , Area Under Curve , Berberine Alkaloids/metabolism , Biological Availability , Blood Proteins/metabolism , Chlorogenic Acid/metabolism , Dose-Response Relationship, Drug , Gastrointestinal Agents/administration & dosage , Gastrointestinal Agents/pharmacokinetics , Injections, Intravenous , Male , Plant Preparations/administration & dosage , Rats, Sprague-Dawley
2.
Int J Clin Pharmacol Ther ; 51(9): 718-24, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23863317

ABSTRACT

Hydrochlorothiazide (HCTZ) is used to treat uncomplicated hypertension. However, many studies have reported the variance of inter-individual response to HCTZ. A meta-analysis of published data was conducted to evaluate the pharmacogenetic associations of ACE I/D and ADD1 Gly460Trp polymorphisms with blood pressure changes during HCTZ therapy. To analyze the influence of ACE I/D polymorphism, 4 studies including 1,439 patients were assessed and the 3 genotypes were compared (II vs. ID, II vs. DD, and ID vs. DD) with respect to blood pressure changes. A significant association between ACE and blood pressure change was observed for the comparison of the II and DD (standard differences in means = 0.256; 95% CI, 0.109 - 0.403). For ADD1 Gly460Trp polymorphism, 4 studies including 1,001 patients were assessed, and GlyGly vs. GlyTrp, GlyGly vs. TrpTrp and GlyTrp vs. TrpTrp genotype comparisons were analyzed. A significant association between ADD1 and blood pressure change was observed for the comparisons of GlyGly vs. GlyTrp (standard differences in means= 2.78; 95% CI, 0.563 - 4.99) and GlyGly vs. TrpTrp (standard differences in means = 1.80; 95% CI, 1.38 - 2.22). This study is the first meta-analysis to evaluate the influences of ACE and ADD1 polymorphisms on blood pressure responses to HCTZ to combine the inconsistent results of previous studies.


Subject(s)
Antihypertensive Agents/pharmacology , Blood Pressure/drug effects , Calmodulin-Binding Proteins/genetics , Hydrochlorothiazide/pharmacology , Peptidyl-Dipeptidase A/genetics , Polymorphism, Genetic , Genotype , Humans , Publication Bias
3.
Xenobiotica ; 43(10): 901-7, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23574017

ABSTRACT

1. Since the prevalent hormonal combination therapy with estrogen analogues in cancer patients has frequency and possibility to induce the cholestasis, the frequent combination therapy with 17α-ethynylestradiol (EE, an oral contraceptive) and doxorubicin (an anticancer drug) might be monitored in aspect of efficacy and safety. Doxorubicin is mainly excreted into the bile via P-glycoprotein (P-gp) and multidrug resistance-associated protein 2 (Mrp2) in hepatobiliary route and metabolized via cytochrome P450 (CYP) 3A subfamily. Also the hepatic Mrp2 (not P-gp) and CYP3A subfamily levels were reduced in EE-induced cholestatic (EEC) rats. Thus, we herein report the pharmacokinetic changes of doxorubicin with respect to the changes in its biliary excretion and hepatic metabolism in EEC rats. 2. The pharmacokinetic study of doxorubicin after intravenous administration of its hydrochloride was conducted along with the investigation of bile flow rate and hepatobiliary excretion of doxorubicin in control and EEC rats. 3. The significantly greater AUC (58.7% increase) of doxorubicin in EEC rats was due to the slower CL (32.9% decrease). The slower CL was due to the reduction of hepatic biliary excretion (67.0% decrease) and hepatic CYP3A subfamily-mediated metabolism (21.9% decrease) of doxorubicin. These results might have broader implications to understand the altered pharmacokinetics and/or pharmacologic effects of doxorubicin via biliary excretion and hepatic metabolism in experimental and clinical estrogen-induced cholestasis.


Subject(s)
Bile/metabolism , Cholestasis/chemically induced , Doxorubicin/pharmacokinetics , Ethinyl Estradiol/adverse effects , Liver/metabolism , Administration, Intravenous , Animals , Antibiotics, Antineoplastic/pharmacokinetics , Area Under Curve , Cholestasis/metabolism , Cytochrome P-450 CYP3A/metabolism , Doxorubicin/administration & dosage , Doxorubicin/blood , Doxorubicin/metabolism , Drug Interactions , Ethinyl Estradiol/pharmacokinetics , Female , Liver/drug effects , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Rats , Rats, Sprague-Dawley
4.
Anticancer Res ; 33(2): 521-8, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23393344

ABSTRACT

Tamoxifen, which is used to treat breast cancer, and ondansetron, used for the treatment of chemotherapy-induced nausea, are commonly metabolized via cytochrome P450 (CYP) 2D subfamily and 3A1/2 in rats, as in humans. This study was conducted to investigate the pharmacokinetic interactions between ondansetron and tamoxifen after intravenous and oral administration of ondansetron (both 8 mg/kg) and/or tamoxifen (2 and 10 mg/kg for intravenous and oral administration, respectively), in rats bearing 7,12-dimethylbenz[a]anthracene (DMBA)-induced mammarian tumors (DMBA rats), used as an animal model of human breast cancer. The total area under the plasma concentration-time curve, from time zero to infinity (AUC) of tamoxifen was significantly greater after both intravenous and oral administration with ondansetron, compared to that after administration of tamoxifen-alone. The hepatic and intestinal metabolism of tamoxifen in DMBA rats was inhibited by ondansetron. Taken together, the significant increase in tamoxifen AUC in DMBA rats after intravenous or oral administration with ondansetron may be attributed to non-competitive hepatic (intravenous) and competitive intestinal (oral) inhibition of CYP2D subfamily- and 3A1/2-mediated tamoxifen metabolism by ondansetron.


Subject(s)
Antiemetics/pharmacokinetics , Mammary Neoplasms, Experimental/chemically induced , Ondansetron/pharmacokinetics , Selective Estrogen Receptor Modulators/pharmacokinetics , Tamoxifen/pharmacokinetics , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Administration, Oral , Animals , Antiemetics/administration & dosage , Antiemetics/metabolism , Area Under Curve , Carcinogens/toxicity , Drug Interactions , Female , Infusions, Intravenous , Mammary Neoplasms, Experimental/drug therapy , Ondansetron/administration & dosage , Ondansetron/metabolism , Rats , Rats, Sprague-Dawley , Selective Estrogen Receptor Modulators/administration & dosage , Selective Estrogen Receptor Modulators/metabolism , Tamoxifen/administration & dosage , Tamoxifen/metabolism
5.
Oncol Rep ; 29(3): 1215-23, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23254912

ABSTRACT

Etoposide, used for the treatment of breast cancer, is mainly metabolized via hepatic cytochrome P450 (CYP) 3A4 in humans and is also a substrate for p-glycoprotein (P-gp). Morin is known to be able to modulate the activities of metabolic enzymes including CYPs and can act as a potent P-gp inhibitor. The purpose of this study was to investigate the effects of morin on the pharmacokinetics of etoposide in rats with 7,12-dimethylbenz[a]anthracene (DMBA)-induced mammary tumors. Etoposide was administered intravenously (2 mg/kg) and orally (10 mg/kg) in control and DMBA rats without (DMBA-WOM) and with (DMBA-WM) morin (15 mg/kg). Protein and mRNA expression of CYP3A and P-gp was analyzed, and the tissue distribution of etoposide was also measured. Both protein and mRNA expression of CYP3A and P-gp was inhibited by morin in the liver, intestine and breast tumors of DMBA-WM rats. After both intravenous and oral administration of etoposide in DMBA-WM rats, the total area under the plasma concentration-time curve from time zero to infinity (AUC) of etoposide was significantly greater, and the time-averaged total body clearance (CL) of etoposide was significantly slower than those in control and DMBA-WOM rats. The amount of etoposide recovered from each tissue was significantly higher in DMBA-WM rats, especially in the breast tumor, liver and large intestine. No significant differences between control and DMBA-WOM rats were observed. Taken together, greater AUC and slower CL of etoposide in DMBA-WM rats could possibly be due to the inhibition of hepatic CYP3A (intravenous) and mainly due to the inhibition of intestinal CYP3A and P-gp (oral) by morin.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents, Phytogenic/pharmacokinetics , Etoposide/pharmacokinetics , Flavonoids/pharmacology , Mammary Neoplasms, Experimental/drug therapy , 9,10-Dimethyl-1,2-benzanthracene , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Adenocarcinoma/chemically induced , Adenocarcinoma/metabolism , Administration, Intravenous , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Cytochrome P-450 CYP3A/genetics , Cytochrome P-450 CYP3A/metabolism , Drug Screening Assays, Antitumor , Etoposide/administration & dosage , Female , Gene Expression , Liver/enzymology , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/metabolism , Rats , Rats, Sprague-Dawley , Tissue Distribution/drug effects
6.
Xenobiotica ; 42(12): 1225-34, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23035955

ABSTRACT

Protein-calorie malnutrition (PCM) could occur frequently in cancer patients and alter the pharmacokinetics of drugs. Also cysteine shows anti-oxidative effect and changes the activities of drug metabolizing enzyme and/or transporters. Herein, we investigated the effects of cysteine on the pharmacokinetics of tamoxifen in rats with protein-calorie malnutrition (PCM). The in vivo pharmacokinetics and in vitro hepatic/intestinal metabolism of tamoxifen were assessed using control, CC (control with cysteine), PCM, PCMC (PCM with cysteine) rats. The effects of cysteine on the intestinal absorption of tamoxifen were further investigated through in vitro transport studies using rat intestine. The AUCs of intravenous tamoxifen in PCM rats were significantly greater than control rats due to the decrease in the hepatic metabolism via CYP3A. In PCMC rats, the elevated AUCs in PCM rats returned to control levels by oral cysteine supplement. The AUC of oral tamoxifen in PCM rats was significantly smaller than in control rats mainly due to the decrease in gastrointestinal absorption. In CC and PCMC rats, oral cysteine supplement enhanced the gastrointestinal absorption of tamoxifen probably via intestinal P-gp inhibition. The present study demonstrated that PCM state and/or oral cysteine supplement had a profound impact on the pharmacokinetics of tamoxifen in rats. If the present rat data are extrapolated to humans, the alterations in tamoxifen absorption and metabolism should be considered in designing a dosage regimen for cancer patients with PCM and/or oral cysteine supplement.


Subject(s)
Cysteine/therapeutic use , Protein-Energy Malnutrition/drug therapy , Tamoxifen/pharmacokinetics , Administration, Intravenous , Administration, Oral , Animals , Bile/metabolism , Biological Transport/drug effects , Blood Proteins/metabolism , Cysteine/pharmacology , Dialysis , Energy Intake/drug effects , Feeding Behavior/drug effects , Intestinal Absorption/drug effects , Intestinal Mucosa/metabolism , Intestines/drug effects , Kinetics , Male , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Protein Binding/drug effects , Protein-Energy Malnutrition/blood , Rats , Rats, Sprague-Dawley , Tamoxifen/administration & dosage , Tamoxifen/blood , Tamoxifen/pharmacology , Weight Gain/drug effects
7.
J Pharm Pharmacol ; 64(10): 1452-60, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22943176

ABSTRACT

OBJECTIVES: Anti-ulcer drugs are frequently used in patients with acute renal failure (ARF). Zinc acexamate is ionized to zinc and ε-acetamidocaproic acid and free EACA exerts a potent therapeutic effect in treating gastric or duodenal ulcers with few side effects. Thus, pharmacokinetic changes in rats with acute renal failure induced by uranyl nitrate (U-ARF rats) were investigated in this study. METHODS: The in-vivo pharmacokinetics and in-vitro hepatic/intestinal metabolism of EACA were assessed using control and U-ARF rats. The mechanism of urinary excretion of EACA was further investigated in rats. KEY FINDINGS: After intravenous and oral administration of zinc acexamate to U-ARF rats, there were significant increases in the values of the area under the curve (AUC) and decreases in the values for time-averaged renal and nonrenal clearances (Cl(r) and Cl(nr) , respectively) compared with control rats. Slower Cl(nr) was partly due to a decrease in the metabolism in liver and/or intestine. Slower Cl(r) could have been due to urine flow rate-dependent timed-interval renal clearance, decrease in organic anion transporter-mediated renal excretion (drug interaction with probenecid and decrease in the relative contribution of net secretion compared with glomerular filtration in U-ARF rats) and/or impaired kidney function. CONCLUSIONS: The pharmacokinetics were significantly altered in U-ARF rats due to the changes in both the hepatic/intestinal metabolism and urinary excretion.


Subject(s)
Acute Kidney Injury/physiopathology , Aminocaproates , Aminocaproic Acid/pharmacokinetics , Intestinal Mucosa/metabolism , Liver/metabolism , Administration, Oral , Aminocaproic Acid/administration & dosage , Animals , Area Under Curve , Disease Models, Animal , Glomerular Filtration Rate , Infusions, Intravenous , Male , Organic Anion Transporters/metabolism , Rats , Rats, Sprague-Dawley , Time Factors , Uranyl Nitrate/toxicity
8.
Xenobiotica ; 42(10): 1017-27, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22568632

ABSTRACT

1. The occurrence of cerebral ischemia is prevalent in patients with hypertension and the combination drug therapy is needed. Thus, the pharmacokinetic interaction between metoprolol (anti-hypertension drug) and SP-8203 (a new drug candidate for cerebral ischemia) with respect to the metabolism via CYP isozymes was evaluated. 2. Metoprolol and SP-8203 were administered intravenously or orally to rats. Concentrations (I) of each drug in the liver and intestine in in vivo studies, the disappearance and apparent K(i) of each drug in in vitro microsomes and [I]/K(i) ratios for each drug were determined. In addition, the disappearance of each drug via CYPs in rat and human microsomes were measured. 3. The AUC and CL(NR) of intravenously administered metoprolol with SP-8203 were significantly greater and slower, respectively, than without SP-8203. However, pharmacokinetic parameters of oral metoprolol and intravenous/oral SP-8203 were not altered. 4. The hepatic metabolism of metoprolol via CYP2D was inhibited by SP-8203 in a competitive manner. However, the intestinal metabolism of metoprolol was not influenced by SP-8203. SP-8203 was not metabolized via CYP isozymes in rats and then co-administration of metoprolol did not affect the metabolism of SP-8203.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Liver/enzymology , Metoprolol/metabolism , Metoprolol/pharmacokinetics , Quinazolinones/pharmacokinetics , Acetamides , Administration, Intravenous , Administration, Oral , Animals , Baculoviridae , Dialysis , Drug Interactions , Humans , Intestinal Mucosa/metabolism , Kinetics , Liver/blood supply , Male , Metoprolol/administration & dosage , Metoprolol/chemistry , Microsomes, Liver/metabolism , Quinazolinones/administration & dosage , Quinazolinones/chemistry , Quinazolinones/metabolism , Rats , Rats, Sprague-Dawley , Regional Blood Flow , Time Factors
9.
Xenobiotica ; 42(8): 766-74, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22300394

ABSTRACT

In this study, the pharmacokinetics of verapamil and its active metabolite norverapamil were evaluated following intravenous and oral administration of 10 mg/kg verapamil to rats with hyperlipidaemia (HL) induced by poloxamer 407 (HL rats). The total area under the plasma concentration time curve (AUC) of verapamil in HL rats following intravenous administration was significantly greater (by 11.2%) than in control rats due to their slower (by 11%) non-renal clearance. The oral AUC of verapamil in HL rats was also significantly greater (by 116%) compared with controls, with a larger magnitude than the data observed following intravenous administration. This may have been a result of the decreased intestinal metabolism of verapamil in HL rats. The AUC of norverapamil and AUC(norverapamil)/AUC(verapamil) ratios following intravenous and oral administration of verapamil were unchanged in HL rats. Assuming that the HL rat model qualitatively reflects similar changes in patients with HL, the findings of this study have potential therapeutic implications. Further studies in humans are required to determine whether modification of the oral verapamil dosage regimen in HL states is necessary.


Subject(s)
Hyperlipidemias/pathology , Verapamil/analogs & derivatives , Administration, Oral , Animals , Blood Proteins/metabolism , Hyperlipidemias/blood , Hyperlipidemias/chemically induced , Infusions, Intravenous , Male , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Poloxamer , Protein Binding/drug effects , Rats , Rats, Sprague-Dawley , Verapamil/administration & dosage , Verapamil/blood , Verapamil/pharmacokinetics
10.
Xenobiotica ; 42(5): 442-55, 2012 May.
Article in English | MEDLINE | ID: mdl-22067009

ABSTRACT

The objective of this study is to report the effects of cysteine on the pharmacokinetics of intravenous and oral docetaxel in rats with protein-calorie malnutrition (PCM). The in vivo pharmacokinetics and in vitro hepatic/intestinal metabolism of docetaxel were assessed using control, CC (control with cysteine), PCM and PCMC (PCM with cysteine) rats. The effects of cysteine on the intestinal absorption of docetaxel were further investigated through in vitro transport studies using rat intestine and Caco-2 cell monolayers. The AUCs (the areas under the plasma concentration-time curve from time zero to time infinity) of intravenous docetaxel in PCM rats were significantly greater than in the control rats because of the significant decrease in the hepatic CYP3A. In PCMC rats, the elevated AUCs in PCM rats returned to control levels. The AUC(0-6 h)s of oral docetaxel in PCM rats were significantly smaller than that in the control rats, mainly due to the decrease in gastrointestinal absorption. In CC and PCMC rats, oral cysteine supplement enhanced the gastrointestinal absorption of docetaxel probably via intestinal P-gp inhibition. If the present rat data could be expressed to humans, the alterations in docetaxel absorption and metabolism should be considered in designing a dosage regimen for cancer patients with PCM state after cysteine supplement.


Subject(s)
Cysteine/therapeutic use , Protein-Energy Malnutrition/drug therapy , Taxoids/pharmacokinetics , Administration, Oral , Animals , Biological Transport/drug effects , Blood Chemical Analysis , Blood Proteins/metabolism , Caco-2 Cells , Docetaxel , Dose-Response Relationship, Drug , Duodenum/drug effects , Duodenum/metabolism , Energy Intake/drug effects , Humans , Injections, Intravenous , Kidney/drug effects , Kidney/pathology , Kinetics , Liver/drug effects , Liver/pathology , Male , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Organ Size/drug effects , Protein Binding/drug effects , Protein-Energy Malnutrition/blood , Protein-Energy Malnutrition/urine , Rats , Rats, Sprague-Dawley , Rhodamine 123/metabolism , Taxoids/administration & dosage , Taxoids/pharmacology , Time Factors , Weight Gain/drug effects
11.
Eur Neuropsychopharmacol ; 22(6): 431-40, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22137858

ABSTRACT

The pharmacokinetics of carbamazepine (CBZ) and its active 10,11-epoxide metabolite (CBZ-E) were evaluated after intravenous and oral administration of 5 mg/kg CBZ to rats with hyperlipidemia induced by poloxamer 407 (HL rats) and controls. The total area under the plasma concentration-time curve (AUC) of CBZ in HL rats after intravenous administration was significantly greater than that in controls due to their slower non-renal clearance (CL(NR)). This was due to slower hepatic CL(int) for metabolism of CBZ to CBZ-E in HL rats via CYP3A1/2. This result was consistent with a previous study indicating reduced hepatic CYP3A1/2 expression in HL rats. Interestingly, the AUC of CBZ-E was also increased in HL rats, while AUC(CBZ-E)/AUC(CBZ) ratios remained unchanged. These results suggested that further metabolism of CBZ-E to the inactive metabolite trans-10,11-dihydoxyl-10,11-dihydro-CBZ (CBZ-D) via microsomal epoxide hydrolase (mEH) was also slowed in HL rats. The significantly reduced hepatic mRNA level and expression of mEH protein in HL rats compared to controls confirmed the above hypothesis. Similar pharmacokinetic changes were observed in HL rats after oral administration of CBZ. These findings have potential therapeutic implications assuming that the HL rat model qualitatively reflects similar changes in patients with hyperlipidemia. Caution is required regarding pharmacotherapy in the hyperlipidemic state in cases where drugs that are metabolized principally by CYP3A1/2 or mEH and have a narrow therapeutic range are in use.


Subject(s)
Analgesics, Non-Narcotic/pharmacokinetics , Carbamazepine/pharmacokinetics , Cytochrome P-450 CYP3A/metabolism , Epoxide Hydrolases/metabolism , Heterocyclic Compounds, 4 or More Rings/metabolism , Thiophenes/metabolism , Analgesics, Non-Narcotic/blood , Animals , Arabidopsis Proteins , Area Under Curve , Carbamazepine/blood , Chromatography, High Pressure Liquid , Disease Models, Animal , Drug Interactions , Epoxide Hydrolases/genetics , Hyperlipidemias/chemically induced , Intramolecular Transferases , Male , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Poloxamer/toxicity , Rats , Rats, Sprague-Dawley , Surface-Active Agents/toxicity , Time Factors
12.
Arch Pharm Res ; 34(11): 1779-88, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22139680

ABSTRACT

Reliably predicting pharmacokinetic behavior in humans from preclinical data is an important aspect of drug development. The most widely used technique in this regard is allometric scaling. In this review, various approaches developed for predicting pharmacokinetic parameters in humans using interspecies scaling are introduced and discussed. Methods to predict plasma concentration-time profiles in humans after intravenous and oral administration are also reviewed. The reliable prediction of human pharmacokinetics with regard to investigational drugs is aimed, ultimately, at selecting the first in-human dose with which to begin clinical studies. Approaches for the selection of the first in-human dose are also reviewed. Although there have been many trials to compare and optimize interspecies scaling methods, no firm conclusions have been reached. Because interspecies scaling methods are still highly empirical, further effort is needed to improve the reliability of predicting human pharmacokinetics by interspecies scaling.


Subject(s)
Drugs, Investigational/pharmacokinetics , Models, Biological , Animals , Body Size , Clinical Trials as Topic/methods , Drug Evaluation, Preclinical/methods , Drug Industry/methods , Drugs, Investigational/analysis , Humans , Species Specificity
13.
Br J Nutr ; 105(2): 220-7, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20819240

ABSTRACT

Astaxanthin is a carotenoid with antioxidant, anti-cancer and anti-inflammatory properties. The pharmacokinetics of astaxanthin after its intravenous (5, 10, and 20 mg/kg) and oral (100 and 200 mg/kg) administration and its first-pass extraction ratios after its intravenous, intraportal or intragastric (20 mg/kg) administration were evaluated in rats. The pharmacokinetic parameters of astaxanthin were dose dependent after its intravenous administration, due to the saturable hepatic metabolism of astaxanthin, but dose independent after oral administration. The gastrointestinal absorption of astaxanthin followed the flip-flop model. The hepatic and gastrointestinal first-pass extraction ratios of astaxanthin were approximately 0·490 and 0·901, respectively. Astaxanthin was metabolised primarily by hepatic cytochrome P-450 1A1/2 in rats. Astaxanthin was unstable up to 4 h incubation in four rat gastric juices and up to 24 h incubation in various buffer solutions having a pH of 1-13. The tissue/plasma ratios of astaxanthin at 8 and 24 h after its oral administration (100 mg/kg) were greater than unity for all tissues studied, except in the heart, at 8 h, indicating that the rat tissues studied had high affinity for astaxanthin.


Subject(s)
Administration, Oral , Animals , Dose-Response Relationship, Drug , Drug Stability , Gastric Mucosa/metabolism , Injections, Intravenous , Liver/metabolism , Male , Methylcholanthrene/pharmacology , Portal Vein , Rats , Rats, Sprague-Dawley , Tissue Distribution , Xanthophylls/administration & dosage , Xanthophylls/metabolism , Xanthophylls/pharmacokinetics
14.
Arch Pharm Res ; 33(7): 1005-8, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20661709

ABSTRACT

Metadoxine [an ion-pair between pyridoxine and pyrrolidone carboxylate (PCA)] plus garlic oil treatment synergistically reduces alcoholic steatosis compared to each agent alone. We evaluated the effect of garlic oil on the pharmacokinetics of pyridoxine. After the oral administration of metadoxine, the total area under the plasma concentration-time curve from time zero to time infinity (AUC) and the peak plasma concentration (C(max)) of pyridoxine were significantly greater (by 40.6%) and higher (by 63.9%), respectively, than after oral administration of pyridoxine plus PCA. Oral metadoxine plus garlic oil also gave larger AUC (31.8%) and higher C(max) (64.9%) than pyridoxine plus PCA. However, garlic oil did not change the AUC or C(max) of pyridoxine in metadoxine. Thus, garlic oil does not enhance the metadoxine activity by affecting the absorption of pyridoxine.


Subject(s)
Allyl Compounds/administration & dosage , Allyl Compounds/pharmacokinetics , Pyridoxine/administration & dosage , Pyridoxine/pharmacokinetics , Pyrrolidonecarboxylic Acid/administration & dosage , Pyrrolidonecarboxylic Acid/pharmacokinetics , Sulfides/administration & dosage , Sulfides/pharmacokinetics , Absorption/drug effects , Absorption/physiology , Administration, Oral , Animals , Drug Combinations , Drug Interactions/physiology , Male , Rats , Rats, Sprague-Dawley
15.
J Pharm Pharm Sci ; 13(1): 93-106, 2010.
Article in English | MEDLINE | ID: mdl-20456834

ABSTRACT

PURPOSE: This study was performed to find which types of hepatic CYP isoforms are responsible for the metabolism of mirodenafil (a new erectogenic) and one of its metabolite, SK3541, using various hepatic CYP inducers and inhibitors in rats. METHODS: Mirodenafil at a dose of 20 mg/kg was administered intravenously to control rats and rats pretreated with various CYP inducers and inhibitors. The disappearance of SK3541 was also measured in vitro hepatic microsomes of rats with and without CYP inducer and inhibitors. RESULTS: Compared to controls, in rats pretreated with 3-methylcholanthrene, orphenadrine, and dexamethasone (main inducers of CYP1A1/2, 2B1/2, and 3A1/2, respectively), the non-renal clearances (CLNRs) of mirodenafil were significantly faster (by 39.4%, 59.3%, and 63.9%, respectively). However, compared to controls, in rats pretreated with quinine and troleandomycin (main inhibitors of CYP2D subfamily and 3A1/2, respectively), the CLNRs of mirodenafil were significantly slower (by 36.1% and 33.2%, respectively). In rat hepatic microsomes spiked with furafylline, quinine, and troleandomycin (main inhibitors of CYP1A2, 2D subfamily, and 3A1/2, respectively), the intrinsic clearances (CLints) for the disappearance of SK3541 were significantly slower (by 18.4%, 35.3%, and 51.5%, respectively) than controls. Also in rat hepatic microsomes pretreated with orphenadrine (a main inducer of CYP2B1/2), the CLint for the disappearance of SK3541 was significantly faster (by 55.5%) than controls. CONCLUSIONS: The above data suggest that hepatic CYP1A1/2, 2B1/2, 2D subfamily, and 3A1/2 are involved in the metabolism of both mirodenafil and SK3541 in rats.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Pyrimidinones/metabolism , Sulfonamides/metabolism , Animals , Enzyme Induction , Enzyme Inhibitors/pharmacology , Male , Microsomes, Liver/metabolism , Phosphodiesterase 5 Inhibitors , Rats , Rats, Sprague-Dawley
16.
Cancer Chemother Pharmacol ; 65(3): 407-18, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19504095

ABSTRACT

PURPOSE: Tamoxifen and ondansetron were commonly metabolized via rat hepatic CYP2D subfamily and 3A1/2, and ondansetron is used to treat chemotherapy-induced nausea. The purpose of this study was to report the pharmacokinetic interaction between tamoxifen and ondansetron in rats. METHODS: The pharmacokinetics of tamoxifen and ondansetron were evaluated after the intravenous and oral administration of tamoxifen, ondansetron, and both drugs together to rats. The Vmax (maximum velocity), Km (apparent Michaelis-Menten constant), CLint (intrinsic clearance), Ki (inhibition constant), and [I] (concentration of inhibitor in the liver and intestine)/Ki ratio of ondansetron were also measured. RESULTS: The AUC0-infinitys of tamoxifen were significantly greater after both intravenous and oral administration with ondansetron compared to those of tamoxifen alone. The significantly slower hepatic and intestinal CLints for the disappearance of tamoxifen with both drugs together were due to inhibition of metabolism of tamoxifen by ondansetron via CYP2D subfamily and 3A1/2. CONCLUSIONS: The significantly greater AUC0-infinity of tamoxifen after the intravenous administration of both drugs together could have possibly been attributable to a non-competitive (hepatic) inhibition of CYP2D subfamily- and 3A1/2-mediated tamoxifen metabolism by ondansetron. The significantly greater AUC0-infinity of tamoxifen after the oral administration of both drugs together could have been attributable to a competitive (intestinal) inhibition of CYP2D subfamily- and 3A1/2-mediated tamoxifen metabolism by ondansetron in addition to non-competitive inhibition in the liver.


Subject(s)
Alcohol Oxidoreductases/metabolism , Aryl Hydrocarbon Hydroxylases/metabolism , Membrane Proteins/metabolism , Ondansetron/pharmacokinetics , Tamoxifen/pharmacokinetics , Administration, Oral , Animals , Area Under Curve , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP3A , Cytochrome P450 Family 2 , Drug Interactions , Infusions, Intravenous , Intestinal Mucosa/metabolism , Kinetics , Liver/metabolism , Male , Metabolic Clearance Rate , Microsomes/metabolism , Ondansetron/administration & dosage , Ondansetron/metabolism , Rats , Rats, Sprague-Dawley , Tamoxifen/administration & dosage , Tamoxifen/metabolism
17.
Pharmacol Ther ; 124(1): 31-43, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19563826

ABSTRACT

Comprehensive studies support the notion that oltipraz [4-methyl-5-(2-pyrazynyl)-1,2-dithiole-3-thione] and its congeners exert cancer chemopreventive effects by the prevention, inhibition or reversal of carcinogenic processes. Recently, it was found that dithiolethione compounds had the activities to prevent or treat fibrosis, insulin resistance, and mitochondrial protective effects in the liver by a mechanism involving AMP-activated protein kinase (AMPK) and/or 70-kDa ribosomal protein S6 kinase 1 (S6K1). Moreover, chemical regulation of the AMPK-S6K1 pathway was found to affect Liver X receptor (LXR) activity and lipogenesis, leading to the identification of AMPK and S6K1 as targets for treating hepatic steatosis. These biological activities of dithiolethiones may offer a novel approach to pharmaceutical intervention. This review focuses on the interaction between oltipraz and the AMPK-mTOR-S6K1 pathway, which regulates genes that confer hepatocyte protection from intoxication, disrupted energy metabolism, and inflammation. In terms of therapeutic potential, the findings reviewed here demonstrate a new therapeutic potential for dithiolethiones, which function in a unique manner, and offer the possibility of new treatments for hepatic diseases.


Subject(s)
Liver Diseases/drug therapy , Pyrazines/therapeutic use , Animals , CCAAT-Enhancer-Binding Protein-beta/metabolism , Fatty Liver/prevention & control , Glutathione Transferase/physiology , Humans , Inactivation, Metabolic , Insulin Resistance , Liver/drug effects , Liver Cirrhosis/prevention & control , MAP Kinase Signaling System , Phosphatidylinositol 3-Kinases/physiology , Protein Kinases/physiology , Pyrazines/pharmacokinetics , Pyrazines/pharmacology , Receptors, Aryl Hydrocarbon/drug effects , Ribosomal Protein S6 Kinases, 70-kDa/physiology , Signal Transduction , TOR Serine-Threonine Kinases , Thiones , Thiophenes
18.
Free Radic Biol Med ; 47(7): 1082-92, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19616619

ABSTRACT

Iron-overload disorders cause hepatocyte injury and inflammation by oxidative stress, possibly leading to liver fibrosis and hepatocellular carcinoma. This study investigated the efficacy of sauchinone, a bioactive lignan, in preventing iron-induced liver injury and explored the mechanism of sauchinone's activity. To create iron overload, mice were injected with phenylhydrazine, and the effects on hepatic iron and histopathology were assessed. Phenylhydrazine treatment promoted liver iron accumulation and ferritin expression, causing hepatocyte death and increased plasma arachidonic acid (AA). Sauchinone attenuated liver injury (EC(50)=10 mg/kg) and activated AMPK in mice. Treatment of hepatocytes with iron and AA simulated iron overload conditions: iron + AA synergistically amplified cytotoxicity, increasing H(2)O(2) and the mitochondrial permeability transition. Sauchinone protected hepatocytes from iron + AA-induced cytotoxicity, preventing the induction of mitochondrial dysfunction and apoptosis (EC(50)=1 microM), similar to the result using metformin. Sauchinone treatment activated LKB1, which led to AMPK activation: these events contributed to cell survival. Evidence of cytoprotection by LKB1 and AMPK activation was revealed in the reversal of sauchinone's restoration of the mitochondrial membrane potential by either dominant negative mutant AMPKalpha or chemical inhibitor. In conclusion, sauchinone protects the liver from toxicity induced by iron accumulation, and sauchinone's effects may be mediated by LKB1-dependent AMPK activation.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Benzopyrans/pharmacology , Dioxoles/pharmacology , Iron/toxicity , Lignans/pharmacology , Liver/drug effects , Liver/injuries , Oxidative Stress/drug effects , Animals , Arachidonic Acid/pharmacology , Benzopyrans/chemistry , Dioxoles/chemistry , Enzyme Activation/drug effects , Iron/administration & dosage , Lignans/chemistry , Liver/pathology , Male , Mice , Mice, Inbred ICR , Phenylhydrazines/pharmacology , Saururaceae/chemistry
19.
J Pharm Pharm Sci ; 12(1): 1-16, 2009.
Article in English | MEDLINE | ID: mdl-19470289

ABSTRACT

PURPOSE: To evaluate the pharmacokinetic interaction between oltipraz and silymarin after intravenous and oral administration of both drugs to male Sprague-Dawley rats. METHODS: Oltipraz (single doses of 10 and 30 mg/kg for intravenous and oral administration, respectively), silymarin (single doses of 50 and 100 mg/kg for intravenous and oral administration, respectively, and 14 days oral administration of 100 mg/kg), alone and together were administered to control rats. RESULTS: The pharmacokinetic parameters of oltipraz did not significantly altered by silymarin. However, after intravenous administration of the drugs together, the AUCs of unconjugated, conjugated, and total (unconjugated plus conjugated) silibinin were significantly different (32.7% decrease, and 32.1% and 27.2% increase, respectively), and total and (CL) and non-renal (CL NR ) clearance of unconjugated silibinin were significantly faster (49.4% and 61.1% increase, respectively) than those of silymarin alone (without oltipraz). After oral administration of silymarin with or without oltipraz, however, the pharmacokinetic parameters of unconjugated, conjugated, and total silibinin were comparable. CONCLUSIONS: After single intravenous administration of the drugs together, the AUC of unconjugated silibinin was significantly smaller, but that of both conjugated and total silibinin was significantly greater. This could have been due to an increase in the formation of conjugates (glucuronidation and sulfation) of silibinin as induced by oltipraz. After simultaneous oral administration of the drugs, however, the AUCs (or AUC 0-12 h) of unconjugated, conjugated, and total silibinin were comparable.


Subject(s)
Pyrazines/pharmacokinetics , Rats, Sprague-Dawley , Silymarin/pharmacokinetics , Administration, Oral , Animals , Anticarcinogenic Agents , Area Under Curve , Drug Interactions , Injections, Intravenous , Male , Metabolic Clearance Rate , Microsomes, Liver/metabolism , Pyrazines/administration & dosage , Pyrazines/pharmacology , Rats , Silybin , Silymarin/administration & dosage , Silymarin/pharmacology , Spectrometry, Mass, Electrospray Ionization , Spectrophotometry, Ultraviolet , Thiones , Thiophenes , Tissue Distribution
20.
Am J Physiol Gastrointest Liver Physiol ; 296(2): G372-81, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19074639

ABSTRACT

Liquiritigenin (LQ), an active component of licorice, has an inhibitory effect on LPS-induced inhibitory nitric oxide synthase expression. This study investigated the effects of LQ on choleresis, the expression of hepatic transporters and phase-II enzymes, and fulminant hepatitis. The choleretic effect and the pharmacokinetics of LQ and its glucuronides were monitored in rats. After intravenous administration of LQ, the total area under the plasma concentration-time curve of glucuronyl metabolites was greater than that of LQ in plasma, which accompanied elevations in bile flow rate and biliary excretion of bile acid, glutathione, and bilirubin. The expressions of hepatocellular transporters and phase-II enzymes were assessed by immunoblots, real-time PCR, and immunohistochemistry. In the livers of rats treated with LQ, the protein and mRNA levels of multidrug resistance protein 2 and bile salt export pump were increased in the liver, which was verified by their increased localizations in canalicular membrane. In addition, LQ treatment enhanced the expression levels of major hepatic phase-II enzymes. Consistent with these results, LQ treatments attenuated galactosamine/LPS-induced hepatitis in rats, as supported by decreases in the plasma alanine aminotransferase, liver necrosis, and plasma TNF-alpha. These results demonstrate that LQ has a choleretic effect and the ability to induce transporters and phase-II enzymes in the liver, which may be associated with a hepatoprotective effect against galactosamine/LPS. Our findings may provide insight into understanding the action of LQ and its therapeutic use for liver disease.


Subject(s)
Bile/metabolism , Carrier Proteins/drug effects , Cholagogues and Choleretics/pharmacology , Enzymes/metabolism , Flavanones/pharmacology , Liver/drug effects , ATP Binding Cassette Transporter, Subfamily B/drug effects , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 11 , ATP-Binding Cassette Transporters/drug effects , ATP-Binding Cassette Transporters/metabolism , Animals , Biotransformation , Carrier Proteins/genetics , Carrier Proteins/metabolism , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/prevention & control , Cholagogues and Choleretics/administration & dosage , Cholagogues and Choleretics/pharmacokinetics , Disease Models, Animal , Enzymes/genetics , Epoxide Hydrolases/metabolism , Flavanones/administration & dosage , Flavanones/pharmacokinetics , Galactosamine , Glucuronides/metabolism , Glucuronosyltransferase/metabolism , Glutathione Transferase/metabolism , Heme Oxygenase (Decyclizing)/metabolism , Infusions, Intravenous , Isoenzymes/metabolism , Lipopolysaccharides , Liver/enzymology , Male , Metabolic Detoxication, Phase II , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Time Factors , Up-Regulation , ATP-Binding Cassette Sub-Family B Member 4
SELECTION OF CITATIONS
SEARCH DETAIL
...