Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Antimicrob Agents Chemother ; 65(8): e0257120, 2021 07 16.
Article in English | MEDLINE | ID: mdl-34097484

ABSTRACT

Delamanid has been studied extensively and approved for the treatment of pulmonary multidrug-resistant tuberculosis; however, its potential in the treatment of extrapulmonary tuberculosis remains unknown. We previously reported that, in rats, delamanid was broadly distributed to various tissues in addition to the lungs. In this study, we simulated human plasma concentration-time courses (pharmacokinetic profile) of delamanid, which has a unique property of metabolism by albumin, using two different approaches (steady-state concentration of plasma-mean residence time [Css-MRT] and physiologically based pharmacokinetic [PBPK] modeling). In Css-MRT, allometric scaling predicted the distribution volume at steady state based on data from mice, rats, and dogs. Total clearance was predicted by in vitro-in vivo extrapolation using a scaled albumin amount. A simulated human pharmacokinetic profile using a combination of human-predicted Css and MRT was almost identical to the observed profile after single oral administration, which suggests that the pharmacokinetic profile of delamanid could be predicted by allometric scaling from these animals and metabolic capacity in vitro. The PBPK model was constructed on the assumption that delamanid was metabolized by albumin in circulating plasma and tissues, to which the simulated pharmacokinetic profile was consistent. Moreover, the PBPK modeling approach demonstrated that the simulated concentrations of delamanid at steady state in the lung, brain, liver, and heart were higher than the in vivo effective concentration for Mycobacterium tuberculosis. These results indicate that delamanid may achieve similar concentrations in various organs to that of the lung and may have the potential to treat extrapulmonary tuberculosis.


Subject(s)
Nitroimidazoles , Tuberculosis , Animals , Antitubercular Agents/therapeutic use , Dogs , Humans , Mice , Models, Biological , Oxazoles , Rats , Tuberculosis/drug therapy
2.
Drug Metab Pharmacokinet ; 39: 100401, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34089983

ABSTRACT

The objective of this study was to obtain the indicators of physicochemical parameters and structurally active sites to design new chemical entities with desirable pharmacokinetic profiles by investigating the process by which machine learning prediction models arrive at their decisions, which are called explainable artificial intelligence. First, we developed the prediction models for metabolic stability, CYP inhibition, and P-gp and BCRP substrate recognition using 265 physicochemical parameters for designing the molecular structures. Four important parameters, including the well-known indicator h_logD, are common in some in vitro studies; as such, these can be used to optimize compounds simultaneously to address multiple pharmacokinetic concerns. Next, we developed machine learning models that had been programmed to show structurally active sites. Many types of machine learning models were developed using the results of in vitro metabolic stability study of around 30000 in-house compounds. The metabolic sites of in-house compounds predicted using some prediction models matched experimentally identified metabolically active sites, with a ratio of number of metabolic sites (predicted/actual) of over 90%. These models can be applied to several screening projects. These two approaches can be employed for obtaining lead compounds with desirable pharmacokinetic profiles efficiently.


Subject(s)
Computer Simulation , Cytochrome P-450 Enzyme Inhibitors , Machine Learning , Artificial Intelligence , Cytochrome P-450 Enzyme Inhibitors/metabolism , Cytochrome P-450 Enzyme Inhibitors/pharmacokinetics , Drug Design/methods , Drug Discovery/methods , Humans , Models, Molecular , Molecular Structure , Predictive Value of Tests , Quantitative Structure-Activity Relationship
3.
Drug Metab Pharmacokinet ; 39: 100395, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33991751

ABSTRACT

We constructed machine learning-based pharmacokinetic prediction models with very high performance. The models were trained on 26138 and 16613 compounds involved in metabolic stability and cytochrome P450 inhibition, respectively. Because the compound features largely differed between the publicly available and in-house compounds, the models learned on the public data could not predict the in-house compounds, suggesting that outside of a certain applicability domain (AD), the prediction results are unreliable and can mislead the design of novel compounds. To exclude the uncertain prediction results, we constructed another machine learning model that determines whether the newly designed compound is inside or outside the AD. The AD was evaluated multi-dimensionally with some explanatory variables: The structural similarities and the probability obtained from the pharmacokinetic prediction model. The accuracy of predicting metabolic stability was 79.9% on the test set, increasing significantly to 93.6% after excluding the low-reliability compounds. The model properly classified the reliability of the compounds. After learning on the in-house compounds, the reliability model classified almost all (90%) of the public compounds as low reliability, indicating that the AD was properly determined by the model.


Subject(s)
Computer Simulation , Drug Discovery/methods , Machine Learning , Pharmaceutical Preparations , Pharmacokinetics , Cytochrome P-450 Enzyme System/metabolism , Humans , Inactivation, Metabolic , Metabolic Clearance Rate/physiology , Pharmaceutical Preparations/analysis , Pharmaceutical Preparations/chemistry , Pharmaceutical Preparations/metabolism , Predictive Value of Tests , Quantitative Structure-Activity Relationship , Reproducibility of Results
4.
Xenobiotica ; 51(5): 590-604, 2021 May.
Article in English | MEDLINE | ID: mdl-33685346

ABSTRACT

The pharmacokinetics of brexpiprazole were investigated in the in vitro and in vivo.The total body clearance of brexpiprazole in rat and monkey was 2.32 and 0.326 L/h/kg, respectively, after intravenous administration, and oral availability was 13.6% and 31.0%, respectively. Dose-dependent exposures were observed at dose ranges between 1-30 mg/kg in the rat and 0.1-3 mg/kg in the monkey.Brexpiprazole distributed widely to body tissues, and Vd,z were 2.81 and 1.82 L/kg in rat and monkey, respectively. The serum protein binding of brexpiprazole was 99% or more in animals and human. Uniform distribution character among the species was suggested by a traditional animal scale-up method.A common main metabolite, DM-3411 was found in animals and humans in the metabolic reactions with the liver S9 fraction. CYP3A4 and CYP2D6 were predominantly involved in the metabolism.The affinity of DM-3411 for D2 receptors was lower than that of brexpiprazole, and neither DM-3411 nor any metabolites with affinity other than M3 were detected in the brain, demonstrating that brexpiprazole is only involved in the pharmacological effects.Overall, brexpiprazole has a simple pharmacokinetic profile with good metabolic stability, linear kinetics, and no remarkable species differences with regard to metabolism and tissue distribution.


Subject(s)
Dopamine , Quinolones , Animals , Haplorhini , Humans , Rats , Serotonin , Thiophenes
5.
Xenobiotica ; 51(5): 522-535, 2021 May.
Article in English | MEDLINE | ID: mdl-33663326

ABSTRACT

Brexpiprazole, a serotonin-dopamine activity modulator, is indicated for the treatment of schizophrenia and also adjunctive therapy to antidepressants for the treatment of Major Depressive Disorder. To determine the drug-drug interaction risk for cytochrome P450, and SLC and ABC transporters, brexpiprazole and its metabolite, DM-3411 were assessed in this in vitro investigation.Brexpiprazole exhibited weak inhibitory effects (IC50 >13 µmol/L) on CYP2C9, CYP2C19, CYP2D6 and CYP3A4 activities, but had moderate inhibitor activity on CYP2B6 (IC50 8.19 µmol/L). The ratio of systemic unbound concentration (3.8 nmol/L) to the Ki value was sufficiently low. DM-3411 had comparable inhibitory potentials with brexpiprazole only for CYP2D6 and CYP3A4. The mRNA expressions of CYP1A2, CYP2B6 and CYP3A4 were not changed by the exposure of brexpiprazole to human hepatocytes.Brexpiprazole and DM-3411 exhibited weak or no inhibitory effects for hepatic and renal transporters (OATPs, OATs, OCTs, MATE1, and BSEP), except for MATE-2K (0.156 µmol/L of DM-3411), even for which the ratio to systemic unbound concentration (5.3 nmol/L) was sufficiently low.Brexpiprazole effected the functions of P-gp and BCRP with IC50 values of 6.31 and 1.16 µmol/L, respectively, however, the pharmacokinetic alteration was not observed in the clinical concomitant study on P-gp and BCRP substrates.These in vitro data suggest that brexpiprazole is unlikely to cause clinically relevant drug interactions resulting from the effects on CYPs or transporters mediating the absorption, metabolism, and/or disposition of co-administered drugs.


Subject(s)
Depressive Disorder, Major , Pharmaceutical Preparations , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Dopamine , Drug Interactions , Humans , Neoplasm Proteins , Quinolones , Serotonin , Thiophenes
6.
Biopharm Drug Dispos ; 38(4): 301-312, 2017 May.
Article in English | MEDLINE | ID: mdl-28092695

ABSTRACT

Delamanid (OPC-67683, Deltyba™, nitro-dihydro-imidazooxazoles derivative) is approved for the treatment of adult pulmonary multidrug-resistant tuberculosis. The absorption, distribution and excretion of delamanid-derived radioactivity were investigated after a single oral administration of 14 C-delamanid at 3 mg/kg to rats. In both male and female rats, radioactivity in blood and all tissues reached peak levels by 8 or 24 h post-dose, and thereafter decreased slowly. Radioactivity levels were 3- to 5-fold higher in lung tissue at time to maximum concentration compared with plasma. In addition, radioactivity was broadly distributed in various tissues, including the central nervous system, eyeball, placenta and fetus, indicating that 14 C-delamanid permeated the brain, retinal and placental blood barriers. By 168 h post-dose, radioactivity in almost all the tissues was higher than that in the plasma. Radioactivity was also transferred into the milk of lactating rats. Approximately 6% and 92% of radioactivity was excreted in the urine and feces, respectively, indicating that the absorbed radioactivity was primarily excreted via the biliary route. No significant differences in the absorption, distribution and excretion of 14 C-delamanid were observed between male and female rats. The pharmacokinetic results suggested that delamanid was broadly distributed to the lungs and various tissues for a prolonged duration of time at concentrations expected to effectively target tuberculosis bacteria. These data indicate that delamanid, in addition to its previously demonstrated efficacy in pulmonary tuberculosis, might be an effective therapeutic approach to treating extrapulmonary tuberculosis. Copyright © 2017 John Wiley & Sons, Ltd.


Subject(s)
Antitubercular Agents/pharmacokinetics , Antitubercular Agents/therapeutic use , Nitroimidazoles/pharmacokinetics , Nitroimidazoles/therapeutic use , Oxazoles/pharmacokinetics , Oxazoles/therapeutic use , Tuberculosis/drug therapy , Animals , Antitubercular Agents/urine , Bile/chemistry , Bile/metabolism , Feces/chemistry , Female , Intestinal Absorption , Liver/metabolism , Male , Maternal-Fetal Exchange , Milk/chemistry , Nitroimidazoles/urine , Oxazoles/urine , Placenta/metabolism , Pregnancy , Rats , Rats, Sprague-Dawley , Tissue Distribution
8.
Antimicrob Agents Chemother ; 60(6): 3497-508, 2016 06.
Article in English | MEDLINE | ID: mdl-27021329

ABSTRACT

Delamanid (Deltyba, OPC-67683) is the first approved drug in a novel class of nitro-dihydro-imidazooxazoles developed for the treatment of multidrug-resistant tuberculosis. Patients with tuberculosis require treatment with multiple drugs, several of which have known drug-drug interactions. Transporters regulate drug absorption, distribution, and excretion; therefore, the inhibition of transport by one agent may alter the pharmacokinetics of another, leading to unexpected adverse events. Therefore, it is important to understand how delamanid affects transport activity. In the present study, the potencies of delamanid and its main metabolites as the substrates and inhibitors of various transporters were evaluated in vitro Delamanid was not transported by the efflux ATP-binding cassette (ABC) transporters P-glycoprotein (P-gp; MDR1/ABCB1) and breast cancer resistance protein (BCRP/ABCG2), solute carrier (SLC) transporters, organic anion-transporting polypeptides, or organic cation transporter 1. Similarly, metabolite 1 (M1) was not a substrate for any of these transporters except P-gp. Delamanid showed no inhibitory effect on ABC transporters MDR1, BCRP, and bile salt export pump (BSEP; ABCB11), SLC transporters, or organic anion transporters. M1 and M2 inhibited P-gp- and BCRP-mediated transport but did so only at the 50% inhibitory concentrations (M1, 4.65 and 5.71 µmol/liter, respectively; M2, 7.80 and 6.02 µmol/liter, respectively), well above the corresponding maximum concentration in plasma values observed following the administration of multiple doses in clinical trials. M3 and M4 did not affect the activities of any of the transporters tested. These in vitro data suggest that delamanid is unlikely to have clinically relevant interactions with drugs for which absorption and disposition are mediated by this group of transporters.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , ATP-Binding Cassette Transporters/metabolism , Antitubercular Agents/pharmacology , Neoplasm Proteins/metabolism , Nitroimidazoles/pharmacology , Organic Anion Transporters/metabolism , Oxazoles/pharmacology , Solute Carrier Proteins/metabolism , Tuberculosis, Multidrug-Resistant/drug therapy , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 11 , Animals , Biological Transport, Active/drug effects , Cell Line , Drug Interactions/physiology , HEK293 Cells , Humans , Kidney Tubules, Proximal/cytology , Nitroimidazoles/metabolism , Octamer Transcription Factor-1/metabolism , Oxazoles/metabolism , Swine
9.
J Pharmacol Sci ; 129(1): 18-25, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26320673

ABSTRACT

Recent studies have shown that rebamipide, which suppresses reactive oxygen species, prevents chemoradiotherapy-induced oral mucositis in patients with head and neck cancers. However, anticancer action of radiotherapy and chemotherapy is believed to be partially associated with generation of reactive oxygen species. The aim of this study was to determine whether rebamipide interferes with the antitumor action of radiotherapy and chemotherapy. The effect of rebamipide on tumor cell growth was investigated using a human oral squamous carcinoma cell line, HSC-2, in vitro and in vivo. Rebamipide showed no significant effect on cell or tumor growth in HSC-2 tumor-bearing nude mice. Influences of rebamipide on the antitumor action of radiotherapy and of chemotherapy with cisplatin or docetaxel were investigated using the same animal model. In radiotherapy, the tumor was treated with 2.5 Gy of X-rays for 5 days, and rebamipide (300 mg/kg p.o.) was administered during irradiation periods. In chemotherapy, tumor-bearing mice were treated once with cisplatin (8 mg/kg, i.v.) or docetaxel (15 mg/kg i.v.) and rebamipide (300 mg/kg p.o.) was administered for 5 days following the antitumor drug treatment. Rebamipide did not interfere with the antitumor action of radiotherapy and chemotherapy.


Subject(s)
Alanine/analogs & derivatives , Antineoplastic Agents/therapeutic use , Antioxidants/pharmacology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/radiotherapy , Cisplatin/therapeutic use , Quinolones/pharmacology , Taxoids/therapeutic use , Alanine/administration & dosage , Alanine/pharmacology , Alanine/therapeutic use , Animals , Antineoplastic Agents/adverse effects , Antioxidants/administration & dosage , Antioxidants/therapeutic use , Cell Line, Tumor , Cisplatin/adverse effects , Disease Models, Animal , Docetaxel , Drug Interactions , Female , Humans , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Quinolones/administration & dosage , Quinolones/therapeutic use , Radiotherapy/adverse effects , Stomatitis/etiology , Stomatitis/prevention & control , Taxoids/adverse effects
10.
Appl Opt ; 51(27): 6653-60, 2012 Sep 20.
Article in English | MEDLINE | ID: mdl-23033038

ABSTRACT

Although some azo-carbazole derivatives attached on or doped into inert polymers are known to show photorefractive effect without external electric field, the origin of their asymmetric energy transfer in two-beam coupling experiments were unknown. We made the two-beam coupling experiment followed by sample translation and one-beam diffraction at 633 nm for thick films composed of 3-[(4-nitrophenyl)]azo-9H-carbazole-9-ethanol (NACzEtOH) and poly(methylmethacrylate), finding that photoinduced gratings grew in several minutes accompanied with phase displacement of the gratings, but the phase shift was not always synchronized with the refractive index modulation. We reformulated the Kogelnik's coupled-wave theory with strict energy conservation law for analysis. Comparison of the grating growth and erasure at 532 nm to Disperse Red 1 (DR1), the most well-known azo dye showed that the photoisomerization was dominant at this wavelength and that the azo-carbazole dyes were faster in response time and more resistive to erasure than DR1.

11.
J Pharm Sci ; 98(5): 1775-87, 2009 May.
Article in English | MEDLINE | ID: mdl-18781650

ABSTRACT

The purpose of the present study was to examine whether the intestinal absorption of low-permeability drugs could be improved by utilization of the intestinal influx transporter PEPT1. We investigated whether peptide derivatives of poorly absorbable nonamino acid-like drugs might be substrates of PEPT1, using rebamipide (Reb) as a model drug. We synthesized several peptide derivatives of rebamipide and examined their inhibitory effect on the uptake of [(3)H]Gly-Sar by PEPT1-expressing HeLa cells. Some of the peptide derivatives inhibited PEPT1-mediated uptake of [(3)H]Gly-Sar. Next, uptake of the inhibitory peptide derivatives was evaluated in PEPT1-expressing Xenopus oocytes and HeLa cells. Ser(Reb)-Gly exhibited significantly increased uptake by PEPT1-expressing cells in comparison with that by mock cells. The permeability of Ser(Reb)-Gly across a Caco-2 cell monolayer was significantly higher than that of rebamipide itself, and the transport was decreased in the presence of PEPT1 substrates. Further, a rat intestinal perfusion study revealed increased absorption of Ser(Reb)-Gly compared with rebamipide. These results demonstrate that the addition of a dipeptide moiety to a poorly absorbable nonpeptide/nonamino acid-like drug can result in absorption via the intestinal transporter PEPT1, though there is some selectivity as regards the structure of the added peptide moiety.


Subject(s)
Alanine/analogs & derivatives , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacokinetics , Intestinal Absorption/physiology , Peptides/chemistry , Quinolones/administration & dosage , Quinolones/pharmacokinetics , Symporters/metabolism , Alanine/administration & dosage , Alanine/pharmacokinetics , Animals , Biological Availability , Caco-2 Cells , Chromatography, High Pressure Liquid , Dipeptides/chemistry , Glutamine/chemistry , HeLa Cells , Humans , In Vitro Techniques , Indicators and Reagents , Intestinal Mucosa/metabolism , Intestines/blood supply , Oocytes/metabolism , Patch-Clamp Techniques , Peptide Transporter 1 , Peptides/chemical synthesis , Rats , Regional Blood Flow , Sodium Dodecyl Sulfate , Taurine , Xenopus
12.
Cancer Sci ; 98(9): 1358-67, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17640296

ABSTRACT

The understanding of the cell signaling pathways and the molecular events leading to cell death of cancer cells will provide in-depth perspective into the identification and development of potent anticancer agents. A balance between cell proliferation and cell death has been raised as a rational target for the management of malignant tumors. In the present study, the authors demonstrated that chemically synthesized sugar-cholestanols consisting of GlcNAcbeta-, Galbeta- and GlcNAcbeta1,3Galbeta-cholestanols exerted a strong inhibiting activity against cell proliferation of esophageal cancer cells, but cholestanol itself did not show such an activity against the same cancer cells at all. In addition to their predominant role as an antiproliferation agent, evidence based on the molecular analyses suggested that sugar-cholestanols played a regulatory role in multiple signal transduction pathways inducing apoptosis through both the death signal-extrinsic and the mitochondria-intrinsic pathways. Sugar-cholestanols seemed to be more susceptible to esophageal cancer cells than to non-cancerous esophageal cells at the very early event of their exposure and, further, to suppress specifically the expression of vascular endothelial growth factor. Taken together, these novel functions of sugar-cholestanols indicate that they could have promising therapeutic potential against human esophageal cancer.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/drug therapy , Cholestanols/chemical synthesis , Cholestanols/pharmacology , Esophageal Neoplasms/drug therapy , Oligosaccharides/chemical synthesis , Oligosaccharides/pharmacology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Caspases/metabolism , Cell Line , Cell Line, Transformed , Cell Line, Tumor , Enzyme Activation , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Growth Inhibitors/chemical synthesis , Growth Inhibitors/pharmacology , Humans , Poly(ADP-ribose) Polymerases/biosynthesis , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Up-Regulation/physiology , Vascular Endothelial Growth Factor A/biosynthesis
13.
Phys Rev Lett ; 96(1): 017404, 2006 Jan 13.
Article in English | MEDLINE | ID: mdl-16486515

ABSTRACT

Solid-state self-quenching processes of highly efficient Ir(III) phosphorescent emitters are investigated by the measurement of thin film photoluminescence quantum efficiency and transient lifetime as a function of doping concentration in a host matrix. The radiative decay rate constant is found to be independent from the average distance between dopant molecules (R), and the concentration-quenching rate constant is shown to be dependent on R(-6). The quenching dependence on R strongly suggests that luminescent concentration quenching in a phosphorescent Ir(III) complex:host film is controlled by dipole-dipole deactivating interactions as described by the Förster energy transfer model.

14.
Drug Metab Pharmacokinet ; 20(6): 491-5, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16415534

ABSTRACT

Grepafloxacin (GPFX) is a new quinolone antibiotic (NQ) which is highly distributed to the lung and other tissues. In the present study, to characterize the distribution mechanism of GPFX to the lung, the uptake of GPFX by isolated rat lung cells was examined in vitro. GPFX was rapidly taken up by the cells, and the uptake reached a steady-state within 5 min. The cell-to-medium concentration ratio at equilibrium was 56.8+/-1.9 microL/mg protein, which was much higher than the cellular volume. GPFX uptake consisted of a saturable component (Km: 264+/-181 microM, Vmax: 2.94+/-2.33 nmol/min/mg protein) and a nonsaturable component (Pdif: 7.04+/-2.17 microL/min/mg protein). The uptake of GPFX was reduced in the presence of ATP-depletors (FCCP and Rotenone) and by the replacement of sodium with choline in the medium, suggesting that GPFX uptake is at least partially mediated by an Na+- and energy-dependent process. GPFX uptake tended to be reduced in the presence of other NQs such as levofloxacin, lomefloxacin and sparfloxacin, but was only minimally affected by the substrates of several uptake mechanisms already identified in the liver and kidney such as taurocholate, p-aminohippurate, L-carnitine and tetraethylammonium. These results suggested that GPFX is taken up by the lung partially via carrier-mediated transport system(s), distinct from the identified transporters, and such active transport systems may at least partially account for the efficient distribution of GPFX to the lung.


Subject(s)
Anti-Bacterial Agents/metabolism , Carrier Proteins/metabolism , Fluoroquinolones/metabolism , Lung/metabolism , Piperazines/metabolism , Adenosine Triphosphate/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Algorithms , Animals , Biological Transport, Active , Kidney/metabolism , Liver/metabolism , Lung/cytology , Male , Rats , Rats, Sprague-Dawley , Sodium/pharmacology , Temperature
15.
Free Radic Res ; 38(5): 487-94, 2004 May.
Article in English | MEDLINE | ID: mdl-15293556

ABSTRACT

Rebamipide, an antiulcer agent, is known as a potent hydroxyl radical (*OH) scavenger. In the present study, we further characterized the scavenging effect of rebamipide against *OH generated by ultraviolet (UV) irradiation of hydrogen peroxide (H2O2), and identified the reaction products to elucidate the mechanism of the reaction. Scavenging effect of rebamipide was accessed by ESR using DMPO as a *OH-trapping agent after UVB exposure (305 nm) to H2O2 for 1 min in the presence of rebamipide. The signal intensity of *OH adduct of DMPO (DMPO-OH) was markedly reduced by rebamipide in a concentration-dependent fashion as well as by dimethyl sulfoxide and glutathione as reference radical scavengers. Their second order rate constant values were 5.62 x 10(10), 8.16 x 10(9) and 1.65 x 10(10) M(-1) s(-1), respectively. As the rebamipide absorption spectrum disappeared during the reaction, a new spectrum grew due to generation of rather specific reaction product. The reaction product was characterized by LC-MS/MS and NMR measurements. Finally, a hydroxylated rebamipide at the 3-position of the 2(1H)-quinolinone nucleus was newly identified as the major product exclusively formed in the reaction between rebamipide and the *OH generated by UVB/H2O2. Specific formation of this product explained the molecular characteristics of rebamipide as a potential *OH scavenger.


Subject(s)
Alanine/analogs & derivatives , Alanine/pharmacology , Anti-Ulcer Agents/pharmacology , Free Radical Scavengers/pharmacology , Hydrogen Peroxide/metabolism , Hydroxyl Radical/chemistry , Quinolones/pharmacology , Chromatography, High Pressure Liquid , Chromatography, Liquid , Cyclic N-Oxides/metabolism , Electron Spin Resonance Spectroscopy , Glutathione/metabolism , Hydroxyl Radical/metabolism , Hydroxylation/drug effects , Hydroxylation/radiation effects , Magnetic Resonance Spectroscopy , Mass Spectrometry , Molecular Structure , Spin Labels , Ultraviolet Rays
16.
J Pharmacol Exp Ther ; 310(2): 648-55, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15131241

ABSTRACT

The involvement of multidrug resistance-associated protein 1 (Mrp1) and P-glycoprotein (mdr1) in the tissue distribution and excretion of grepafloxacin (GPFX), a fluoroquinolone antibiotic, was investigated using gene-deficient mice [mdr1a(-/-), mdr1a/1b(-/-), and mrp1(-/-)]. The plasma concentration-time profile of GPFX in mrp1(-/-) was nearly identical to that in mrp1(+/+), whereas that in mdr1a/1b(-/-) was higher than that in mdr1a/1b(+/+). The urinary clearance of GPFX in mdr1a/1b(-/-) was lower than that in mdr1a/1b(+/+), suggesting that the urinary excretion of GPFX is at least partially mediated by mdr1. The tissue-to-plasma concentration ratios during the beta-phase (K(p beta),) was significantly higher in the heart, trachea, kidney, spleen, and brown fat of mrp1(-/-) than those in mrp1(+/+). In MRP1-transfected LLC-PK1 cells, the efflux of GPFX after preloading into the cells was higher than that observed in the parent cell lines. These results suggest that GPFX is a substrate of MRP1 and that its distribution to these tissues might be limited by Mrp1. On the other hand, a higher K(p beta), and of GPFX in mdr1a(-/-) mdr1a/1b(-/-) compared with mdr1a/1b(+/+) was observed only in the brain. GPFX was efficiently distributed to the lung parenchyma cells and pulmonary airspaces, including the epithelial lining fluid and macrophages that are the pharmacological target of GPFX, although the contribution of Mdr1 and Mrp1 to such distribution seems to be minor. Thus, the present findings reveal that the disposition of GPFX is at least in part governed by these two ABC transporters and that both Mrp1 and Mdr1 are involved in the limited distribution of GPFX to the distinct tissues, including pharmacological and/or toxicological targets by an active efflux mechanism.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/deficiency , Fluoroquinolones/blood , Fluoroquinolones/urine , Piperazines/blood , Piperazines/urine , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Animals , Fluoroquinolones/pharmacology , Male , Mice , Mice, Knockout , Piperazines/pharmacology , Tissue Distribution/drug effects , Tissue Distribution/physiology
17.
Drug Metab Dispos ; 30(12): 1393-9, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12433809

ABSTRACT

This study was carried out to investigate the most important factor(s) governing the tissue distribution of grepafloxacin (GPFX), a fluoroquinolone antibiotic, in rats. The tissue-to-blood concentration ratio (K(p)) of GPFX at steady state during constant infusion was highest in the lung, followed by the pancreas, kidney, and spleen. After bolus injection, GPFX was efficiently taken up by most of the organs examined, the uptake clearance other than the lung being almost blood flow-limited. Approximately 10% of the intravenously injected dose was rapidly trapped by the lung, but GPFX distribution rapidly decreased within 30 s due to the washout by the plasma flow. Thus, the higher distribution of GPFX to the lung compared with the other organs cannot be accounted for by a difference in its uptake or efflux. Subcellular fractionation after the infusion indicated that GPFX is primarily distributed to the organelle fractions in most organs, 60% of lung-associated GPFX being recovered in the nucleus and plasma membrane fraction. Such subcellular distribution in the lung was proportional to the phosphatidylserine (PhS) content of each fraction. The steady-state K(p) value in each tissue in vivo also correlated with the tissue content of PhS. GPFX preferentially binds to PhS, compared with other phospholipids, and this binding was inhibited by weakly basic drugs, such as quinidine, imipramine, and propranolol, that have also been reported to bind to PhS. The association of GPFX with PhS synthase transformants of Chinese hamster ovary (CHO-K1) cells depends on the PhS content of each cell line, this association being also inhibited by basic drugs. These results suggest that binding of GPFX to PhS is the major determinant of the high distribution of GPFX to the lung.


Subject(s)
Anti-Infective Agents/metabolism , Fluoroquinolones , Piperazines/metabolism , Animals , Anti-Infective Agents/chemistry , Area Under Curve , Dose-Response Relationship, Drug , Male , Metabolic Clearance Rate/drug effects , Metabolic Clearance Rate/physiology , Piperazines/chemistry , Rats , Rats, Sprague-Dawley , Tissue Distribution/drug effects , Tissue Distribution/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...