Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 122
Filter
1.
Int J Mol Sci ; 19(4)2018 Apr 09.
Article in English | MEDLINE | ID: mdl-29642532

ABSTRACT

The in vivo relevance of ursodeoxycholate (UDCA) treatment (100 mg/kg/day, per oral tid for 5 days before cholestasis induction followed by the same dosing for 5 days) on hepatic function was investigated in rats with 17α-ethinylestradiol (EE, 10 mg/kg, subcutaneous for 5 days)-induced experimental cholestasis. The bile flow rate and the expression level of hepatic multidrug resistance-associated protein 2 (Mrp 2) that were decreased in cholestasis were restored after UDCA treatment. Consistent with this, the biliary excretion clearance (CLexc,bile) of a representative Mrp2 substrate-methotrexate (MTX)-was decreased in cholestatic rats but was restored after UDCA treatment. Consequently, the plasma concentrations of MTX, which were increased by cholestasis, were decreased to control levels by UDCA treatment. Thus, the restoration of CLexc,bile appears to be associated with the increase in Mrp2 expression on the canalicular membrane by UDCA treatment followed by Mrp2-mediated biliary excretion of MTX. On the other hand, the hepatic uptake clearance (CLup,liver) of MTX was unchanged by cholestasis or UDCA treatment, suggestive of the absence of any association between the uptake process and the overall biliary excretion of MTX. Since UDCA has been known to induce the expression of canalicular MRP2 in humans, UDCA treatment might be effective in humans to maintain or accelerate the hepatobiliary elimination of xenobiotics or metabolic conjugates that are MRP2 substrates.


Subject(s)
Bile Acids and Salts/chemistry , Cholestasis/prevention & control , Ethinyl Estradiol/adverse effects , Methotrexate/blood , Ursodeoxycholic Acid/administration & dosage , ATP-Binding Cassette Transporters/metabolism , Administration, Oral , Animals , Cholestasis/blood , Cholestasis/chemically induced , Cholestasis/metabolism , Down-Regulation , Drug Administration Schedule , Male , Multidrug Resistance-Associated Protein 2 , Rats , Treatment Outcome , Ursodeoxycholic Acid/pharmacology
2.
J Pharm Biomed Anal ; 151: 61-70, 2018 Mar 20.
Article in English | MEDLINE | ID: mdl-29306735

ABSTRACT

We previously reported that MDR-1339, an inhibitor of ß-amyloid protein aggregation, was likely to be eliminated by biotransformation in rats. The objective of this study was to determine the chemical identity of metabolites derived from this aggregate inhibitor and to characterize the kinetics of formation of these metabolites in rats. Using high performance liquid chromatography coupled with mass spectrometry with a hybrid triple quadrupole-linear ion trap, 7 metabolites and 1 potential metabolic intermediate were identified in RLM incubations containing MDR-1339. In addition to these, 3 glucuronide metabolites were detected in urine samples from rats receiving a 10 mg/kg oral dose of MDR-1339. When the kinetics of the formation of two major metabolites, M1 and M2, were analyzed assuming simple Michaelis-Menten kinetics, the Vmax and Km values were found to be 0.459 ±â€¯0.0196 nmol/min/mg protein and 28.3 ±â€¯3.07 µM for M1, and 0.101 ±â€¯0.00537 nmol/min/mg protein and 14.7 ±â€¯2.37 µM for M2, respectively. When chemically synthesized M1 and M2 were individually administered to rats intravenously at the dose of 5 mg/kg respectively, the volume of distribution and elimination clearance were determined to be 4590 ±â€¯709 mL/kg and 68.4 ±â€¯5.60 mL/min/kg for M1 and 15300 ±â€¯8110 mL/kg and 98.0 ±â€¯19.5 mL/min/kg for M2, respectively. When MDR-1339 was intravenously administered to rats at a dose of 5 mg/kg, the parent drug and M1 were readily detected for periods of up to 6 h after the administration, but M2 was observed only from 2 to 4 h. A standard moment analysis indicates that the formation clearance of M1 is 6.01 mL/min/kg, suggesting that 19.7% of the MDR-1339 dose was eliminated in rats. These observations indicate that the hepatic biotransformation of MDR-1339 results in the formation of at least 10 metabolites and that M1 is the major metabolite derived from this aggregation inhibitor in rats.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Benzofurans/metabolism , Microsomes, Liver/metabolism , Protein Aggregates/drug effects , Animals , Benzofurans/pharmacology , Dose-Response Relationship, Drug , Male , Microsomes, Liver/drug effects , Protein Aggregates/physiology , Rats , Rats, Sprague-Dawley
3.
Drug Dev Ind Pharm ; 43(3): 441-447, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27824264

ABSTRACT

A 3-month depot formulation of leuprolide acetate (Luphere 3M Depot) with a mean microsphere diameter of 22.3 µm was prepared aseptically by spray-drying glacial acetic acid solution of the drug and polylactic acid, and lyophilization in a d-mannitol solution. The encapsulation efficiency and loading content of the drug in the Luphere 3M Depot were 94.7% and 9.92% (w/w), respectively. The in vitro release of leuprolide from the depot was substantially delayed and the release profile was similar to that of Lucrin Depot (Abbott Korea, Korea). The safety and pharmacokinetics of leuprolide were investigated over a period of 42 days in 20 prostate cancer patients following a subcutaneous injection of Luphere 3M or Lucrin Depot suspensions (leuprolide acetate dose of 11.25 mg) in a multi-center, randomized, single dose, parallel study. Both formulations were well tolerated by the patients and no serious adverse effects were observed during and after the study. No significant differences were observed in the maximum serum concentration (Cmax) and area under the curve (AUClast) of leuprolide between the two formulations. The results suggest comparable safety and efficacy profiles of Luphere 3M Depot and Lucrin Depot in clinical situations.


Subject(s)
Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/pharmacokinetics , Leuprolide/administration & dosage , Leuprolide/pharmacokinetics , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Aged , Antineoplastic Agents, Hormonal/administration & dosage , Antineoplastic Agents, Hormonal/chemistry , Delayed-Action Preparations/chemistry , Drug Compounding , Humans , Injections, Subcutaneous , Leuprolide/chemistry , Male , Middle Aged , Time Factors
4.
J Colloid Interface Sci ; 480: 102-108, 2016 Oct 15.
Article in English | MEDLINE | ID: mdl-27421112

ABSTRACT

Chitosan (CS)-modified poly(d,l-lactic-co-glycolic) acid (PLGA) nanoparticles (NPs) were prepared and their lung targetability after intravenous administration was elucidated. PLGA NPs (mean diameter: 225nm; polydispersity index: 0.11; zeta potential: -15mV), 0.2% (w/v) CS-coated PLGA NPs (CS02-PLGA NPs, mean diameter: 264nm; polydispersity index: 0.17; zeta potential: -7mV), and 0.5% (w/v) CS-coated PLGA NPs (CS05-PLGA NPs, mean diameter: 338nm; polydispersity index: 0.23; zeta potential: 12mV) were fabricated by a modified solvent evaporation method. PLGA NPs maintained their initial particle size in different media, such as human serum albumin (HSA) solution, rat plasma, and distilled water (DW), while CS05-PLGA NPs exhibited the formation of aggregates in early incubation time and disassembly of those into the NPs in late incubation time (at 24h). According to the sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) analysis, the binding affinity of CS05-PLGA NPs with HSA and rat plasma was higher than that of PLGA NPs. By a near-infrared fluorescence (NIRF) imaging test in the mouse, the selective accumulation of CS05-PLGA NPs, rather than PLGA NPs, in lung tissue was demonstrated. These findings suggest that CS05-PLGA NPs can form transient aggregates in the blood stream after intravenous administration and markedly improve lung targeting efficiency, compared with PLGA NPs.


Subject(s)
Chitosan/chemistry , Lactic Acid/blood , Lactic Acid/pharmacokinetics , Lung/metabolism , Nanoparticles/chemistry , Polyglycolic Acid/pharmacokinetics , Animals , Chitosan/administration & dosage , Chitosan/blood , Chitosan/pharmacokinetics , Female , Humans , Lactic Acid/administration & dosage , Lactic Acid/chemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/metabolism , Particle Size , Polyglycolic Acid/administration & dosage , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Rats , Surface Properties , Tissue Distribution
5.
Drug Des Devel Ther ; 9: 1815-23, 2015.
Article in English | MEDLINE | ID: mdl-25848218

ABSTRACT

BACKGROUND: The identification of permeation enhancers has gained interest in the development of drug delivery systems. A six-mer peptide, H-FCIGRL-OH (AT1002), is a tight junction modulator with promising permeation-enhancing activity. AT1002 enhances the transport of molecular weight markers or agents with low bioavailability with no cytotoxicity. However, AT1002 is not stable in neutral pH or after incubation under physiological conditions, which is necessary to fully uncover its permeation-enhancing effect. Thus, we increased the stability or mitigated the instability of AT1002 by modifying its terminal amino acids and evaluated its subsequent biological activity. METHODS: C-terminal-amidated (FCIGRL-NH2, Pep1) and N-terminal-acetylated (Ac-FCIGRL, Pep2) peptides were analyzed by liquid chromatography-mass spectrometry. We further assessed cytotoxicity on cell monolayers, as well as the permeation-enhancing activity following nasal administration of the paracellular marker mannitol. RESULTS: Pep1 was nontoxic to cell monolayers and showed a relatively low decrease in peak area compared to AT1002. In addition, administration of mannitol with Pep1 resulted in significant increases in the area under the plasma concentration-time curve and peak plasma concentration at 3.63-fold and 2.68-fold, respectively, compared to mannitol alone. In contrast, no increase in mannitol concentration was shown with mannitol/AT1002 or mannitol/Pep2 compared to the control. Thus, Pep1 increased the stability or possibly reduced the instability of AT1002, which resulted in an increased permeation-enhancing effect of AT1002. CONCLUSION: These results suggest the potential usefulness of C-terminal-amidated AT1002 in enhancing nasal drug delivery, which may lead to the development of a practical drug delivery technology for drugs with low bioavailability.


Subject(s)
Amides/chemistry , Drug Delivery Systems , Oligopeptides/administration & dosage , Oligopeptides/pharmacokinetics , Administration, Intranasal , Animals , Biological Availability , Caco-2 Cells , Humans , Hydrogen-Ion Concentration , Male , Nasal Mucosa/drug effects , Oligopeptides/chemistry , Rats , Rats, Sprague-Dawley
6.
J Photochem Photobiol B ; 140: 49-56, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25090224

ABSTRACT

Photodynamic therapy (PDT) in combination with chemotherapy has great potential for cancer treatment. However, there have been very few attempts to developing cancer-targeted co-delivered systems of photosensitizers and anticancer drugs. We developed hematoporphyrin (HP)-modified doxorubicin (DOX)-loaded nanoparticles (HP-NPs) to improve the therapeutic effect of PDT in treating liver cancer. HP is not only a ligand for low density lipoprotein (LDL) receptors on the hepatoma cells but also a well-known photosensitizer for PDT. In vitro phototoxicity in HepG2 (human hepatocellular carcinoma) cells and in vivo anticancer efficacy in HepG2 tumor-bearing mice of free HP and HP-NPs were evaluated. The in vitro phototoxicity in HepG2 cells determined by MTT assay, annexin V-FITC staining and FACS analysis was enhanced in HP-NPs compared with free HP. Furthermore, compared with free HP-based PDT, in vivo anticancer efficacy in HepG2 tumor-bearing mice was markedly improved by HP-NPs-based PDT. Moreover, in both cases, the therapeutic effect was increased according to the irradiation time and number of PDT sessions. In conclusion, the HP-NPs prepared in this study represent a potentially effective co-delivery system of photosensitizer (HP) and anticancer drug (DOX) which improved the effects of PDT in liver cancer.


Subject(s)
Antineoplastic Agents/chemistry , Doxorubicin/chemistry , Hematoporphyrins/chemistry , Nanoparticles/chemistry , Photosensitizing Agents/chemistry , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/radiation effects , Carcinoma, Hepatocellular/drug therapy , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Drug Carriers/chemistry , Hematoporphyrins/pharmacology , Hep G2 Cells , Humans , Light , Liver Neoplasms/drug therapy , Mice , Mice, Inbred BALB C , Mice, Nude , Photochemotherapy , Photosensitizing Agents/pharmacology , Transplantation, Heterologous
7.
Biopharm Drug Dispos ; 35(1): 60-9, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24151179

ABSTRACT

Anti-cancer agents delivered to cancer cells often show multi-drug resistance (MDR) due to expulsion of the agents. One way to address this problem is to increase the accumulation of anti-cancer agents in cells via amino acid transporters. Thus, val-lapatinib and tyr-lapatinib were newly synthesized by adding valine and tyrosine moieties, respectively, to the parent anti-cancer agent lapatinib without stability issues in rat plasma. Val-lapatinib and tyr-lapatinib showed enhanced anti-cancer effects versus the parent lapatinib in various cancer cell lines, including human breast cancer cells (MDA-MB-231, MCF7) and lung cancer cells (A549), but not in non-cancerous MDCK-II cells. A glutamine uptake study revealed that both val-lapatinib and tyr-lapatinib, but not the parent lapatinib, inhibited glutamine transport in MDA-MB-231 and MCF7 cells, suggesting the involvement of amino acid transporters. In conclusion, val-lapatinib and tyr-lapatinib have enhanced anti-cancer effects, likely due to an increased uptake of the agents into cancer cells via amino acid transporters. The present data suggest that amino acid transporters may be an effective drug delivery target to increase the uptake of anti-cancer agents, leading to one method of overcoming MDR in cancer cells.


Subject(s)
Amino Acid Transport Systems/metabolism , Antineoplastic Agents/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Quinazolines/administration & dosage , Tyrosine/chemistry , Valine/chemistry , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Dogs , Glutamine/metabolism , Humans , Lapatinib , Madin Darby Canine Kidney Cells , Protein Kinase Inhibitors/chemistry , Quinazolines/chemistry
8.
Nat Prod Commun ; 8(8): 1143-8, 2013 Aug.
Article in English | MEDLINE | ID: mdl-24079189

ABSTRACT

We demonstrated that an aqueous extract of dried immature fruit of Poncirus trifoliate (PF-W) produces relaxation of intestinal smooth muscle using the ileac strips of a rat. Furthermore, the underlying mechanism of its relaxant activity was investigated. PF-W was prepared using the standard extraction protocol. A 1.5 - 2 cm long rat ileac strip was placed in an organ bath with Tyrode's solution and smooth muscle contractility was recorded by connecting it to a force transducer. Various compounds were added to the organ baths, and changes in muscular contractility were measured. PF-W concentration-dependently induced relaxation of rat ileac strips that were contracted both spontaneously and via acetylcholine treatment. Various potassium channel blockers did not inhibit the relaxation by PF-W. No difference in the effect of PF-W was observed between ileac strips treated with low (20 mM) and high concentrations (60 mM) of KCl. PF-W inhibited the contraction of rat ileac strips induced by extracellular calcium. PF-W acts as a potent smooth muscle relaxant, implicating its possible action as a rapid acting reliever for abdominal pains and a cure for intestinal convulsion. Considering that PF-W also exhibits prokinetic activity, its use in various gastrointestinal disorders seems promising.


Subject(s)
Gastrointestinal Agents/pharmacology , Ileum/drug effects , Muscle, Smooth/drug effects , Plant Extracts/pharmacology , Poncirus/chemistry , Animals , Fruit/chemistry , Gastrointestinal Agents/analysis , Gastrointestinal Motility/drug effects , In Vitro Techniques , Male , Rats , Rats, Sprague-Dawley
9.
Int J Pharm ; 456(2): 362-9, 2013 Nov 18.
Article in English | MEDLINE | ID: mdl-24012861

ABSTRACT

The aim of this study was to investigate the feasibility of transdermal fluoxetine (FX) delivery. The effects of chemical forms (base or salt) and permeation enhancers on in vitro skin permeation of FX were assessed using hairless mouse, rat and human cadaver skin. The optimized formulations from the in vitro studies were then evaluated in an in vivo pharmacokinetic study in rats. The in vitro skin permeation studies suggested that the FX base (FXB) and isopropyl myristate (IPM)-limonene mixture could be suitable for transdermal delivery of FX. The permeation parameters of FX through human cadaver skin were well correlated with that through hairless mouse and rat skin, suggesting that these animal models can be used for predicting the permeability of FX through human skin. After transdermal administration of FX with IPM or the IPM-limonene mixture to rats, the mean steady-state plasma concentration (Css) was 66.20 or 77.55 ng/mL, respectively, which was maintained over 36 h and had a good correlation with the predicted Css from the in vitro data. These in vitro and in vivo data demonstrated that permeation enhancers could be a potential strategy for transdermal delivery of FX.


Subject(s)
Drug Delivery Systems/methods , Fluoxetine/administration & dosage , Fluoxetine/pharmacokinetics , Skin Absorption/drug effects , Administration, Cutaneous , Animals , Dose-Response Relationship, Drug , Humans , Male , Mice , Mice, Hairless , Organ Culture Techniques , Permeability/drug effects , Rats , Rats, Sprague-Dawley , Skin Absorption/physiology
10.
Int J Biol Macromol ; 61: 175-81, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23817101

ABSTRACT

The aim of this study is to evaluate the feasibility of hyaluronic acid/sodium alginate (HA/SA) scaffold-based interpenetrating polymeric network (IPN) for the proliferation and chondrogenic differentiation of the human adipose-derived stem cells (hADSCs). The hADSCs cultured in HA/SA IPN scaffold exhibited enhanced cell adhesion and proliferation compared to the HA scaffold. Superior chondrogenic differentiation of hADSCs in HA/SA IPN scaffold, compared to HA-based scaffold, was confirmed by measuring expression levels of chondrogenic markers. These results suggested that HA/SA IPN scaffold could provide a desirable environment for the cell adhesion, proliferation and chondrogenic differentiation of hADSCs.


Subject(s)
Adipose Tissue/cytology , Alginates/chemistry , Hyaluronic Acid/chemistry , Stem Cells/cytology , Tissue Scaffolds/chemistry , Cell Culture Techniques , Cell Differentiation , Cell Proliferation , Chondrogenesis , Collagen Type II/metabolism , Glucuronic Acid/chemistry , Hexuronic Acids/chemistry , Humans , Immunohistochemistry , Porosity , Stem Cells/metabolism , Stem Cells/ultrastructure , Tissue Engineering/methods
11.
Drug Dev Ind Pharm ; 39(10): 1515-22, 2013 Oct.
Article in English | MEDLINE | ID: mdl-22934579

ABSTRACT

The Biopharmaceutics Classification System (BCS) was developed to facilitate estimation of the in vivo pharmacokinetic performance of drugs from human intestinal permeability and solubility. However, the measurement of human in vivo intestinal permeability, unlike that of solubility, is problematic and inefficient. Thus, rat in vitro intestinal permeability results obtained via the Ussing chamber technique are often used instead. However, these data could be unreliable due to difficulty in maintaining the viability of the dissected intestinal membrane in the Ussing chamber. Therefore, a more efficient method to obtain a reliable in vitro permeability is mandatory. Here, we propose a new approach by introducing a novel factor called the permeability ratio (PR). Basically, PR is a rat in vitro intestinal permeability obtained from the Ussing chamber, which is then corrected by the permeability of lucifer yellow, a paracellular permeability marker. To prove the validity of the method, 12 model drugs representing different BCS classes were tested, and the correlation with human in vivo intestinal permeability was high. More importantly, the new method perfectly classified all 12 model drugs. The results indicate that PR is a reliable factor with high correlation to human in vivo intestinal permeability, which can further be used to accurately predict the BCS classification.


Subject(s)
Drug Evaluation, Preclinical/methods , Intestinal Absorption , Jejunum/metabolism , Models, Biological , Pharmaceutical Preparations/metabolism , Algorithms , Animals , Humans , In Vitro Techniques , Male , Permeability , Pharmaceutical Preparations/analysis , Pharmaceutical Preparations/chemistry , Pharmaceutical Preparations/classification , Pharmacokinetics , Rats , Rats, Sprague-Dawley , Solubility
12.
Int J Pharm ; 441(1-2): 562-9, 2013 Jan 30.
Article in English | MEDLINE | ID: mdl-23142421

ABSTRACT

Because Mycobacterium tuberculosis, which causes tuberculosis, survives mainly in the alveolar macrophages, the remedial efficiency of anti-tuberculosis drugs such as ofloxacin may be improved by their direct delivery to the lungs via pulmonary inhalation. For this purpose, ofloxacin-loaded, glutaraldehyde-crosslinked chitosan microspheres (OCMs) were prepared using a water-in-oil emulsification method. The particle size of the OCMs was around 1-6 µm, and the content of ofloxacin was 27% (w/w). A twin-stage impinger (TSI) study revealed that the device-removal efficiency of the drug from the capsule and the arrival rate of the drug to stage II of the apparatus were substantially improved for OCMs compared to ofloxacin itself (i.e., 81 vs. 98% and 13 vs. 45%, respectively). Also, the in vitro uptake of ofloxacin from the OCMs to alveolar macrophages (NR8383) was substantially accelerated: the cellular ofloxacin concentrations at 4 and 24 h after the application were >3.5-fold greater than those for free ofloxacin. The above results indicate that pulmonary inhalation of OCMs might improve the delivery efficiency of ofloxacin to the alveolar macrophages, thereby shortening the length of time that is required to cure tuberculosis with the drug-usually at least 6 months when administered orally.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Chitosan/chemistry , Drug Delivery Systems , Ofloxacin/pharmacokinetics , Administration, Inhalation , Animals , Anti-Bacterial Agents/administration & dosage , Cross-Linking Reagents/chemistry , Glutaral/chemistry , Macrophages, Alveolar/metabolism , Ofloxacin/administration & dosage , Particle Size , Rats , Time Factors
13.
Int J Pharm ; 441(1-2): 373-80, 2013 Jan 30.
Article in English | MEDLINE | ID: mdl-23174411

ABSTRACT

Chitosan oligosaccharide-arachidic acid (CSOAA) conjugate was successfully synthesized and used for the development of self-assembled nanoparticles for doxorubicin (DOX) delivery. The molar substitution of AA on CSO and critical micelle concentration (CMC) of CSOAA were measured. Physicochemical properties of DOX-loaded CSOAA-based nanoparticles, such as particle size, zeta potential and morphology, were also characterized. The DOX-loaded CSOAA-based nanoparticles showed spherical shape with a mean diameter of 130 nm and positive charge. According to the result of in vitro release test, DOX-loaded CSOAA-based nanoparticles exhibited sustained and pH-dependent drug release profiles. The CSOAA showed negligible cytotoxicity in FaDu, human head and neck cancer, cells. Cellular uptake of DOX in FaDu cells was higher in the nanoparticle-treated group compared to the free DOX group. The anti-tumor efficacy of DOX-loaded nanoparticles was also verified in FaDu tumor xenografted mouse model. These results suggested that synthesized amphiphilic CSOAA might be used for the preparation of self-assembled nanoparticles for anti-cancer drug delivery.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Doxorubicin/pharmacology , Drug Delivery Systems , Head and Neck Neoplasms/drug therapy , Animals , Antibiotics, Antineoplastic/administration & dosage , Cell Line, Tumor , Chitosan/chemistry , Delayed-Action Preparations , Doxorubicin/administration & dosage , Drug Carriers/chemistry , Eicosanoic Acids/chemistry , Female , Head and Neck Neoplasms/pathology , Humans , Hydrogen-Ion Concentration , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles , Oligosaccharides/chemistry , Particle Size , Xenograft Model Antitumor Assays
14.
Xenobiotica ; 43(7): 579-91, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23252722

ABSTRACT

1. Doxorubicin exhibited dose-independent pharmacokinetics after intravenous (5-20 mg/kg) and oral (20-100 mg/kg) administration to rats. Nearly all (82.1-99.7%) of the orally administered doxorubicin remained unabsorbed, and the hepatic first-pass extraction ratio and oral bioavailability of doxorubicin were approximately 0.5% and 1%, respectively. Based on these results, it is likely that the primary factor responsible for the low oral bioavailability of doxorubicin is the limited intestinal absorption, rather than the CYP3A4-mediated first-pass metabolism. 2. Moreover, the in vitro transport and cellular uptake studies using Caco-2 cell monolayers have revealed that doxorubicin crosses the intestinal epithelium primarily via the paracellular pathway (accounting for 85.6% of the overall absorptive transport) probably due to its physicochemical properties (hydrophilic cation; pKa = 9.67, log P = -0.5). These results suggest that P-glycoprotein (P-gp)-mediated efflux activity does not play a significant role in limiting the intestinal absorption of doxorubicin, attenuating the absorptive transport by only 5.56-13.2%. 3. Taken together, the present study demonstrated that the limited and paracellular intestinal absorption of doxorubicin was a major factor responsible for its low oral bioavailability, restricting the role of CYP3A4-mediated first-pass metabolism and P-gp-mediated efflux.


Subject(s)
Antibiotics, Antineoplastic/pharmacokinetics , Doxorubicin/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Administration, Oral , Animals , Biological Availability , Caco-2 Cells , Cytochrome P-450 CYP3A/metabolism , Humans , Intestinal Absorption , Male , Rats , Rats, Sprague-Dawley
15.
Xenobiotica ; 43(4): 355-67, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23020787

ABSTRACT

The oral (po) bioavailability of gemifloxacin mesylate in rats and its possible association with efflux transporters was investigated. The apparent permeabilities (Papp) of gemifloxacin across the Caco-2 cell monolayer were 1.20 ± 0.09 × 10(-5) cm/s for apical to basal (absorptive) transport, and 2.13 ± 0.6 × 10(-5) cm/s for basal to apical (secretory) transport for a 5-500 µM concentration range, suggesting the involvement of a carrier-mediated efflux in the secretory transport. The secretory transport in Caco-2 cells was significantly decreased by MRP2 (MK571) and BCRP (Ko143) inhibitors. The secretory transport was distinct in MDCKII/P-gp, MDCKII/MRP2 and MDCKII/BCRP cells, and the affinity was highest for MRP2, followed by BCRP and P-gp. The efflux was significantly decreased by verapamil and Ko143, but not significantly by MK571. The comparative po bioavailability in rats was increased by the preadministration of Ko143 (four-fold), MK571 (two-fold) and verapamil (two-fold). Efflux transporters appeared to significantly limit the bioavailability of gemifloxacin in rats, suggesting their possible contribution to the low bioavailability of the drug in the human (70%).


Subject(s)
Anti-Bacterial Agents/metabolism , Fluoroquinolones/metabolism , Intestinal Absorption , Membrane Transport Proteins/metabolism , Naphthyridines/metabolism , Quinolones/metabolism , Administration, Oral , Animals , Anti-Bacterial Agents/blood , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacokinetics , Biological Transport/drug effects , Caco-2 Cells , Dogs , Fluoroquinolones/blood , Fluoroquinolones/chemistry , Fluoroquinolones/pharmacokinetics , Gemifloxacin , Humans , Inhibitory Concentration 50 , Kinetics , Madin Darby Canine Kidney Cells , Male , Naphthyridines/blood , Naphthyridines/chemistry , Naphthyridines/pharmacokinetics , Quinolones/blood , Quinolones/chemistry , Quinolones/pharmacokinetics , Rats , Rats, Sprague-Dawley , Verapamil/pharmacology
16.
Arch Pharm Res ; 35(8): 1441-8, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22941487

ABSTRACT

We studied the impact of experimental kidney failure on the pharmacokinetics of a model organic cation and investigated the underlying mechanism(s) of the organic cation transporters. The systemic pharmacokinetics and tissue distribution of triethylmethylammonium (TEMA), a model organic cation, were characterized after intravenous doses of 0.3-30 µmol/kg in rats with or without uranyl nitrate-induced acute renal failure (UN-ARF). To study the effect of endogenous substrates in plasma from UN-ARF rats on organic cation transport, rOCT- or rOCT2-dependent uptake of tetraethylammonium (TEA) was studied in rOCT1-transfected or rOCT2-transfected LLC-PK1 cells, respectively. As a result, the AUC for TEMA was increased, probably because of decreased total clearance, and the tissue-to-plasma concentration ratio (T/P ratio) of TEMA was unchanged in the liver but decreased significantly in the kidneys of UN-ARF rats. In vitro, the uptake of TEA was decreased significantly by adding UN-ARF plasma, compared with control plasma, in rOCT2-overexpressing LLC-PK1 cells, but not in rOCT1-overexpressing LLC-PK1 cells. These observations suggest that the induction of UN-ARF leads to an accumulation of endogenous organic cation(s), probably rOCT2 substrate(s), in the plasma, thereby affecting the TEMA pharmacokinetics and distribution to the kidneys in rats.


Subject(s)
Acute Kidney Injury/physiopathology , Organic Cation Transport Proteins/metabolism , Organic Cation Transporter 1/metabolism , Quaternary Ammonium Compounds/pharmacokinetics , Administration, Intravenous , Animals , Area Under Curve , Dose-Response Relationship, Drug , LLC-PK1 Cells , Liver/metabolism , Male , Organic Cation Transporter 2 , Quaternary Ammonium Compounds/administration & dosage , Rats , Rats, Sprague-Dawley , Swine , Tissue Distribution , Transfection , Uranyl Nitrate/toxicity
17.
Pharm Res ; 29(12): 3443-54, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22886625

ABSTRACT

PURPOSE: Hyaluronic acid-ceramide (HACE)-based nanoparticles (NPs) were developed for the targeted delivery of doxorubicin (DOX), and their antitumor efficacy for melanoma was evaluated. METHODS: DOX-loaded HACE-based self-assembled NPs were prepared and their physicochemical properties were characterized. The in vitro cytotoxicity of HACE was measured using an MTS-based assay. The cellular uptake efficiency of DOX into mouse melanoma B16F10 cells was assessed by confocal laser scanning microscopy and flow cytometry. Tumor growth and body weight were monitored after the intratumoral and intravenous injection of DOX-loaded NPs into a B16F10 tumor-bearing mouse model. RESULTS: DOX-loaded NPs, with a mean diameter of ~110 nm, a narrow size distribution, and high drug entrapment efficiency, were prepared. A sustained DOX release pattern was shown, and drug release was enhanced at pH 5.5 compared with pH 7.4. The cytotoxicity of HACE to B16F10 cells was negligible. It was assumed that DOX was taken up into the B16F10 cells through receptor-mediated endocytosis. A significant inhibitory effect was observed on tumor growth, without any serious changes in body weight, after the injection of DOX-loaded NPs into the B16F10 tumor-bearing mouse model. CONCLUSIONS: DOX-loaded HACE-based NPs were successfully developed and their antitumor efficacy against B16F10 tumors was demonstrated.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Doxorubicin/administration & dosage , Drug Delivery Systems , Melanoma/drug therapy , Nanoparticles/chemistry , Animals , Antibiotics, Antineoplastic/pharmacokinetics , Antibiotics, Antineoplastic/therapeutic use , Cell Line, Tumor , Ceramides/chemistry , Doxorubicin/pharmacokinetics , Doxorubicin/therapeutic use , Female , Hyaluronic Acid/chemistry , Melanoma/pathology , Mice , Mice, Inbred C57BL
18.
Xenobiotica ; 42(11): 1110-9, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22747239

ABSTRACT

Identifying kinetic determinants of hepatic elimination of drugs would be crucial for better understanding its pharmacokinetics and predicting drug interactions. Present study investigated the kinetics of sinusoidal uptake of docetaxel and its impact on the overall hepatic elimination of docetaxel in rats. The non-renal clearance (CL(NR); hepatic elimination) of docetaxel were significantly reduced by co-administration of intravenous rifampicin, a potent inhibitor of organic anion transporting peptides (OATPs; Oatps), at a dose of 20 mg/kg. Docetaxel uptake into isolated rat hepatocytes was found to be temperature/concentration/energy-dependent, saturable, and reduced by Oatps inhibitors (rifampicin and bromosulfophthalein). Moreover, docetaxel uptake into perfused rat liver was significantly reduced in the presence of 10-µM rifampicin. However, docetaxel metabolism in rat hepatic microsome was not affected by rifampicin at less than 50 µM. Based on the comparison of intrinsic clearances related to hepatic clearance, it can be suggested that sinusoidal uptake could be the rate-determining process in the overall hepatic elimination of docetaxel in rats.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Liver/metabolism , Organic Anion Transporters/metabolism , Taxoids/pharmacokinetics , Animals , Docetaxel , Enzyme Inhibitors , Hepatocytes/metabolism , In Vitro Techniques , Kinetics , Male , Metabolic Clearance Rate , Microsomes, Liver/metabolism , Rats , Rats, Sprague-Dawley , Rifampin
19.
Int J Pharm ; 434(1-2): 191-8, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22643227

ABSTRACT

Phytosphingosine and methyl derivatives are important mediators on cellular processes, and are associated with cell growth and death. The antitumor activity of N,N,N-trimethylphytosphingosine-iodide (TMP) as a novel potent inhibitor of angiogenesis and metastasis was evaluated in B16F10 murine melanoma cells. The results indicated that TMP itself effectively inhibited in vitro cell migration, tube formation, and the expression of angiogenic factors as well as in vivo lung metastasis. However, TMP slightly suppressed in vivo experimental tumor metastasis in its free form and induced side effects including hemolysis and local side effects. Therefore, in an attempt to reduce the toxicity and the undesirable side effects of TMP, a liposomal formulation was prepared and tested for its effectiveness. TMP liposomes retained the effectiveness of TMP in vitro while side effects were reduced, and both in vivo experimental and spontaneous tumor metastasis were significantly suppressed. These results support the conclusion that TMP effectively inhibits in vitro angiogenesis as well as in vivo metastasis, and a liposomal formulation is more efficient delivery system for TMP treatment than solution.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Melanoma, Experimental/drug therapy , Quaternary Ammonium Compounds/pharmacology , Sphingosine/analogs & derivatives , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/toxicity , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/toxicity , Cell Line, Tumor , Cell Movement/drug effects , Hemolysis/drug effects , Humans , Liposomes , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Melanoma, Experimental/blood supply , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Quaternary Ammonium Compounds/administration & dosage , Quaternary Ammonium Compounds/toxicity , Rats , Sphingosine/administration & dosage , Sphingosine/pharmacology , Sphingosine/toxicity
20.
Xenobiotica ; 42(7): 603-13, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22233275

ABSTRACT

The phenomenon known as multiple-drug resistance, whereby anti-cancer agents are expelled from cancer cells, makes it necessary to develop methods that will reliably increase the accumulation of anti-cancer agents within cancer cells. To accomplish this goal, a new model compound, Val-SN-38, was synthesized by introducing valine to SN-38, an active ingredient of irinotecan. Val-SN-38 improved intracellular accumulation approximately 5-fold in MCF7 cells, compared with SN-38, and rather than changes in membrane permeability, the amino acid transporter ATB(0,+) played a role, whereas the dipeptide transporter PEPT1 did not. Other sodium-dependent amino acid transporters, namely ATA1, ATA2, and ASCT2, were unexpectedly involved in the uptake of Val-SN-38 as well. The efflux of Val-SN-38 by major efflux transporters was variably changed, but not significantly. In summary, the enhanced accumulation of Val-SN-38 in cancer cells was due to augmented uptake via various amino acid transporters. The results of the present study make a compelling argument in favour of a prodrug concept that can improve intracellular accumulation and take advantage of amino acid transporters without significantly inducing multiple-drug resistance.


Subject(s)
Amino Acid Transport Systems/metabolism , Antineoplastic Agents/metabolism , Camptothecin/analogs & derivatives , Valine/metabolism , Amino Acid Transport System A/metabolism , Amino Acid Transport System ASC/metabolism , Antineoplastic Agents/chemical synthesis , Biological Transport , Camptothecin/chemistry , Camptothecin/metabolism , Esters , HEK293 Cells , Humans , Irinotecan , Minor Histocompatibility Antigens , Prodrugs/chemical synthesis , Prodrugs/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...