Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Molecules ; 23(6)2018 06 17.
Article in English | MEDLINE | ID: mdl-29914211

ABSTRACT

Honokiol (2-(4-hydroxy-3-prop-2-enyl-phenyl)-4-prop-2-enyl-phenol) and magnolol (4-Allyl-2-(5-allyl-2-hydroxy-phenyl)phenol) are the major active polyphenol constituents of Magnolia officinalis (Magnoliaceae) bark, which has been widely used in traditional Chinese medicine (Houpu Tang) for the treatment of various diseases, including anxiety, stress, gastrointestinal disorders, infection, and asthma. The aim of this study was to investigate the direct effects of honokiol and magnolol on hepatic CYP1A and 2C-mediated metabolism in vitro using rat liver microsomes and in vivo using the Sprague-Dawley rat model. Honokiol and magnolol inhibited in vitro CYP1A activity (probe substrate: phenacetin) more potently than CYP2C activity (probe substrate: diclofenac): The mean IC50 values of honokiol for the metabolism of phenacetin and diclofenac were 8.59 µM and 44.7 µM, while those of magnolol were 19.0 µM and 47.3 µM, respectively. Notably, the systemic exposure (AUC and Cmax) of phenacetin, but not of diclofenac, was markedly enhanced by the concurrent administration of intravenous honokiol or magnolol. The differential effects of the two phytochemicals on phenacetin and diclofenac in vivo pharmacokinetics could at least be partly attributed to their lower IC50 values for the inhibition of phenacetin metabolism than for diclofenac metabolism. In addition, the systemic exposure, CL, and Vss of honokiol and magnolol tended to be similar between the rat groups receiving phenacetin and diclofenac. These findings improve our understanding of CYP-mediated drug interactions with M. officinalis and its active constituents.


Subject(s)
Biphenyl Compounds/administration & dosage , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 Enzyme System/metabolism , Diclofenac/pharmacokinetics , Lignans/administration & dosage , Liver/enzymology , Phenacetin/pharmacokinetics , Administration, Intravenous , Animals , Biphenyl Compounds/pharmacology , Chromatography, High Pressure Liquid , Drug Interactions , Gene Expression Regulation/drug effects , Lignans/pharmacology , Liver/cytology , Microsomes, Liver/enzymology , Molecular Structure , Rats , Rats, Sprague-Dawley
2.
PLoS One ; 12(3): e0173247, 2017.
Article in English | MEDLINE | ID: mdl-28273121

ABSTRACT

Carfilzomib (CFZ) is a peptide epoxyketone proteasome inhibitor approved for the treatment of multiple myeloma (MM). Despite the remarkable efficacy of CFZ against MM, the clinical trials in patients with solid cancers yielded rather disappointing results with minimal clinical benefits. Rapid degradation of CFZ in vivo and its poor penetration to tumor sites are considered to be major factors limiting its efficacy against solid cancers. We previously reported that polymer micelles (PMs) composed of biodegradable block copolymers poly(ethylene glycol) (PEG) and poly(caprolactone) (PCL) can improve the metabolic stability of CFZ in vitro. Here, we prepared the CFZ-loaded PM, PEG-PCL-deoxycholic acid (CFZ-PM) and assessed its in vivo anticancer efficacy and pharmacokinetic profiles. Despite in vitro metabolic protection of CFZ, CFZ-PM did not display in vivo anticancer efficacy in mice bearing human lung cancer xenograft (H460) superior to that of the clinically used cyclodextrin-based CFZ (CFZ-CD) formulation. The plasma pharmacokinetic profiles of CFZ-PM were also comparable to those of CFZ-CD and the residual tumors that persisted in xenograft mice receiving CFZ-PM displayed an incomplete proteasome inhibition. In summary, our results showed that despite its favorable in vitro performances, the current CFZ-PM formulation did not improve in vivo anticancer efficacy and accessibility of active CFZ to solid cancer tissues over CFZ-CD. Careful consideration of the current results and potential confounding factors may provide valuable insights into the future efforts to validate the potential of CFZ-based therapy for solid cancer and to develop effective CFZ delivery strategies that can be used to treat solid cancers.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Micelles , Oligopeptides/administration & dosage , Oligopeptides/pharmacokinetics , Polymers , Proteasome Inhibitors/administration & dosage , Proteasome Inhibitors/pharmacokinetics , Animals , Cell Line, Tumor , Disease Models, Animal , Drug Compounding , Drug Design , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Proteasome Endopeptidase Complex/metabolism , Xenograft Model Antitumor Assays
3.
Eur J Pharm Biopharm ; 115: 1-17, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28215648

ABSTRACT

The objective of the current study was to determine the minimum permeability coefficient, P, needed for perfusion-limited distribution in PBPK. Two expanded kinetic models, containing both permeability and perfusion terms for the rate of tissue distribution, were considered: The resulting equations could be simplified to perfusion-limited distribution depending on tissue permeability. Integration plot analyses were carried out with theophylline in 11 typical tissues to determine their apparent distributional clearances and the model-dependent permeabilities of the tissues. Effective surface areas were calculated for 11 tissues from the tissue permeabilities of theophylline and its PAMPA P. Tissue permeabilities of other drugs were then estimated from their PAMPA P and the effective surface area of the tissues. The differences between the observed and predicted concentrations, as expressed by the sum of squared log differences with the present models were at least comparable to or less than the values obtained using the traditional perfusion-limited distribution model for 24 compounds with diverse PAMPA P values. These observations suggest that the use of a combination of the proposed models, PAMPA P and the effective surface area can be used to reasonably predict the pharmacokinetics of 22 out of 24 model compounds, and is potentially applicable to calculating the kinetics for other drugs. Assuming that the fractional distribution parameter of 80% of the perfusion rate is a reasonable threshold for perfusion-limited distribution in PBPK, our theoretical prediction indicates that the pharmacokinetics of drugs having an apparent PAMPA P of 1×10-6cm/s or more will follow the traditional perfusion-limited distribution in PBPK for major tissues in the body.


Subject(s)
Theophylline/pharmacokinetics , Tissue Distribution/physiology , Animals , Male , Models, Biological , Perfusion/methods , Permeability , Pharmacokinetics , Rats , Rats, Sprague-Dawley
4.
J Pharm Biomed Anal ; 137: 155-162, 2017 Apr 15.
Article in English | MEDLINE | ID: mdl-28122294

ABSTRACT

EC-18 (i.e., 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol), an active ingredient in Rockpid®, has been reported to be useful in controlling various types of inflammations, particularly those caused by neutropenia. Although this product was originally approved as a functional food in Korea, it is currently in phase II clinical trials for use in managing the severe chemotherapy-induced neutropenia in patients with advanced breast cancer who are receiving intermediate febrile neutropenia risk chemotherapy. The objective of this study was to develop a rapid, sensitive method for the determination of EC-18 in rat and mouse plasma and to evaluate the applicability of the assay in pharmacokinetic studies. EC-18 was extracted with MeOH from rat and mouse plasma samples, and the extract directly introduced onto an LC-MS/MS system. The analyte and EC-18-d3, an internal standard, were analyzed by multiple reaction monitoring (MRM) at m/z transitions of 635.4→355.4 for EC-18 and 638.4→338.4 for the internal standard, respectively. The lower limit of quantification (LLOQ) was determined at 50ng/mL, with an acceptable linearity in the range from 50 to 10,000ng/mL (r>0.999) for both matrices. Validation parameters such as accuracy, precision, dilution, recovery, matrix effects and stability were found to be within the acceptance criteria of the assay validation guidelines, indicating that the assay is applicable for estimating EC-18 in concentrations in the range examined. EC-18 was readily determined in plasma samples for periods of up to 8h following an intravenous bolus injection of 1mg/kg in rats and at 5mg/kg in mice, respectively, and up to 24h following the oral administration of 2000mg/kg in mice. The findings indicate that the current analytical method is applicable for pharmacokinetic studies of EC-18 in small animals.


Subject(s)
Diglycerides/blood , Diglycerides/chemistry , Glycerol/blood , Glycerol/chemistry , Plasma/chemistry , Animals , Chromatography, Liquid/methods , Drug Stability , Male , Mice , Mice, Inbred ICR , Rats , Rats, Sprague-Dawley , Tandem Mass Spectrometry/methods
5.
Drug Metab Dispos ; 45(3): 246-259, 2017 03.
Article in English | MEDLINE | ID: mdl-28069721

ABSTRACT

Cytochrome P450 enzymes and human organic anion transporting polypeptide (OATP) 1B1 are reported to be involved in the pharmacokinetics of lobeglitazone (LB), a new peroxisome proliferator-activated receptor γ agonist. Atorvastatin (ATV), a substrate for CYP3A and human OATP1B1, is likely to be coadministered with LB in patients with the metabolic syndrome. We report herein on a study of potential interactions between LB and ATV in rats. When LB was administered intravenously with ATV, the systemic clearance and volume of distribution at steady state for LB remained unchanged (2.67 ± 0.63 ml/min per kg and 289 ± 20 ml/kg, respectively), compared with that of LB without ATV (2.34 ± 0.37 ml/min per kg and 271 ± 20 ml/kg, respectively). Although the tissue-to-plasma partition coefficient (Kp) of LB was not affected by ATV in most major tissues, the liver Kp for LB was decreased by ATV coadministration. Steady-state liver Kp values for three levels of LB were significantly decreased as a result of ATV coadministration. LB uptake was inhibited by ATV in rat OATP1B2-overexpressing Madin-Darby canine kidney cells and in isolated rat hepatocytes in vitro. After incorporating the kinetic parameters for the in vitro studies into a physiologically based pharmacokinetics model, the characteristics of LB distribution to the liver were consistent with the findings of the in vivo study. It thus appears that the distribution of LB to the liver is mediated by the hepatic uptake of transporters such as rat OATP1B2, and carrier-mediated transport is involved in the liver-specific drug-drug interaction between LB and ATV in vivo.


Subject(s)
Atorvastatin/pharmacology , Liver/metabolism , Pyrimidines/pharmacokinetics , Solute Carrier Organic Anion Transporter Family Member 1B3/metabolism , Thiazolidinediones/pharmacokinetics , Animals , Atorvastatin/blood , Biological Transport , Dogs , Dose-Response Relationship, Drug , Drug Interactions , Injections, Intravenous , Madin Darby Canine Kidney Cells , Male , Metabolic Clearance Rate , Microsomes, Liver/metabolism , Models, Biological , Pyrimidines/blood , Rats, Sprague-Dawley , Solute Carrier Organic Anion Transporter Family Member 1B3/genetics , Substrate Specificity , Thiazolidinediones/blood , Tissue Distribution , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...