Toshiyuki Kudo
Musashino University
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Toshiyuki Kudo.
Drug Metabolism and Disposition | 2013
Toshiyuki Kudo; Akihiro Hisaka; Yuichi Sugiyama; Kiyomi Ito
The plasma concentration of repaglinide is reported to increase greatly when given after repeated oral administration of itraconazole and gemfibrozil. The present study analyzed this interaction based on a physiologically based pharmacokinetic (PBPK) model incorporating inhibition of the hepatic uptake transporter and metabolic enzymes involved in repaglinide disposition. Firstly, the plasma concentration profiles of inhibitors (itraconazole, gemfibrozil, and gemfibrozil glucuronide) were reproduced by a PBPK model to obtain their pharmacokinetic parameters. The plasma concentration profiles of repaglinide were then analyzed by a PBPK model, together with those of the inhibitors, assuming a competitive inhibition of CYP3A4 by itraconazole, mechanism-based inhibition of CYP2C8 by gemfibrozil glucuronide, and inhibition of organic anion transporting polypeptide (OATP) 1B1 by gemfibrozil and its glucuronide. The plasma concentration profiles of repaglinide were well reproduced by the PBPK model based on the above assumptions, and the optimized values for the inhibition constants (0.0676 nM for itraconazole against CYP3A4; 14.2 μM for gemfibrozil against OATP1B1; and 5.48 μM for gemfibrozil glucuronide against OATP1B1) and the fraction of repaglinide metabolized by CYP2C8 (0.801) were consistent with the reported values. The validity of the obtained parameters was further confirmed by sensitivity analyses and by reproducing the repaglinide concentration increase produced by concomitant gemfibrozil administration at various timings/doses. The present findings suggested that the reported concentration increase of repaglinide, suggestive of synergistic effects of the coadministered inhibitors, can be quantitatively explained by the simultaneous inhibition of the multiple clearance pathways of repaglinide.
Life Sciences | 2011
Nobutomo Ikarashi; Toshihide Mochiduki; Ayaka Takasaki; Takashi Ushiki; Kohta Baba; Makoto Ishii; Toshiyuki Kudo; Kiyomi Ito; Takahiro Toda; Wataru Ochiai; Kiyoshi Sugiyama
AIMS We have suggested that an osmotic laxative, magnesium sulphate (MgSO(4)), may act as a cathartic in a very rational manner by increasing the aquaporin 3 (AQP3) expression level and by changing osmotic pressure in the colon. In this study, we examined the mechanism by which MgSO(4) increases the intestinal AQP3 expression level by using the human colon cancer HT-29 cell line. MAIN METHODS After the addition of MgSO(4) to HT-29 cells, the expression levels of AQP3 mRNA and protein were measured using real-time RT-PCR and western blotting, respectively. The intracellular Mg(2+) concentration, adenylate cyclase (AC) activity and protein kinase A (PKA) activity were also determined. The phosphorylated cAMP response element-binding protein (CREB) expression levels were determined by western blotting. KEY FINDINGS The AQP3 mRNA expression level started to increase significantly at 1 h after MgSO(4) addition and peaked at 9 h, at a level 3 times as high as the control levels. The AQP3 protein expression level started to increase 6 h after the addition and reached a level almost twice as high as the control levels by hour 12. In the HT-29 cells treated with MgSO(4), there was a 1.4-fold increase in the intracellular Mg(2+) concentration, a 1.5-fold increase in AC activity, a 1.6-fold increase in PKA activity, and a significant increase in phosphorylation of the CREB. SIGNIFICANCE These results suggest that the AC activation caused by an increase in the intracellular Mg(2+) concentration may trigger CREB phosphorylation through PKA activation and promote AQP3 gene transcription.
Drug Metabolism and Pharmacokinetics | 2011
Azusa Hoshino-Yoshino; Motohiro Kato; Kohnosuke Nakano; Masaki Ishigai; Toshiyuki Kudo; Kiyomi Ito
The purpose of this study was to provide a pharmacokinetics/pharmacodynamics and toxicokinetics/toxicodynamics bridging of kinase inhibitors by identifying the relationship between their clinical and preclinical (rat, dog, and monkey) data on exposure and efficacy/toxicity. For the eight kinase inhibitors approved in Japan (imatinib, gefitinib, erlotinib, sorafenib, sunitinib, nilotinib, dasatinib, and lapatinib), the human unbound area under the concentration-time curve at steady state (AUC(ss,u)) at the clinical dose correlated well with animal AUC(ss,u) at the no-observed-adverse-effect level (NOAEL) or maximum tolerated dose (MTD). The best correlation was observed for rat AUC(ss,u) at the MTD (p < 0.001). E(max) model analysis was performed using the efficacy of each drug in xenograft mice, and the efficacy at the human AUC of the clinical dose was evaluated. The predicted efficacy at the human AUC of the clinical dose varied from far below E(max) to around E(max) even in the tumor for which use of the drugs had been accepted. These results suggest that rat AUC(ss,u) at the MTD, but not the efficacy in xenograft mice, may be a useful parameter to estimate the human clinical dose of kinase inhibitors, which seems to be currently determined by toxicity rather than efficacy.
Xenobiotica | 2016
Toshiyuki Kudo; Yuya Ozaki; Tomomi Kusano; Eri Hotta; Yuka Oya; Seina Komatsu; Hitomi Goda; Kiyomi Ito
Abstract 1. Buffer conditions in in vitro metabolism studies using human liver microsomes (HLM) have been reported to affect the metabolic activities of several cytochrome P450 (CYP) isozymes in different ways, although there are no reports about the dependence of CYP2C8 activity on buffer conditions. 2. The present study investigated the effect of buffer components (phosphate or Tris-HCl) and their concentration (10–200 mM) on the CYP2C8 and CYP3A4 activities of HLM, using paclitaxel and triazolam, respectively, as marker substrates. 3. The Km (or S50) and Vmax values for both paclitaxel 6α-hydroxylation and triazolam α- and 4-hydroxylation, estimated by fitting analyses based on the Michaelis–Menten or Hill equation, greatly depended on the buffer components and their concentration. 4. The CLint values in phosphate buffer were 1.2–3.0-fold (paclitaxel) or 3.1–6.4-fold (triazolam) higher than in Tris-HCl buffer at 50–100 mM. These values also depended on the buffer concentration, with a maximum 2.3-fold difference observed between 50 and 100 mM which are both commonly used in drug metabolism studies. 5. These findings suggest the necessity for optimization of the buffer conditions in the quantitative evaluation of metabolic clearances, such as in vitro–in vivo extrapolation and also estimating the contribution of a particular enzyme in drug metabolism.
Xenobiotica | 2015
Toshiyuki Kudo; Yumiko Endo; Rina Taguchi; Masami Yatsu; Kiyomi Ito
Abstract 1. Blood levels of S-warfarin have been reported to be increased by concomitant administration of metronidazole (MTZ), an antiprotozoal imidazole derivative. 2. To elucidate the mechanism of this interaction and to identify other possible drug-drug interactions, we conducted an in vitro study with the human hepatoma HepaRG cells and cryopreserved human hepatocytes on the ability of MTZ to reduce the expression of cytochrome P450 (CYP) as well as nuclear receptors that regulate the expression of these enzymes. 3. HepaRG cells and cryopreserved human hepatocytes were treated with MTZ (20 to 500 µM) and were then analyzed by real-time RT-PCR to determine mRNA levels of drug-metabolizing enzymes and nuclear receptors. 4. In both cells, the expressions of CYP2C8, CYP2C9, CYP3A4 and constitutive androstane receptor (CAR) were decreased by MTZ treatment. Particularly, in HepaRG cells, their mRNA levels were decreased by MTZ treatment in a concentration-dependent manner. 5. Our findings suggest that the interaction between MTZ and S-warfarin may be due to the MTZ-induced down-regulation of CYP2C9, the primary enzyme responsible for S-warfarin hydroxylation, and CAR, which regulates CYP2C9 expression. We also found that MTZ use may alter the disposition of drugs metabolized by the CYP isozymes investigated.
International Journal of Pharmaceutics | 2015
Kazuhisa Ozeki; Motohiro Kato; Yuuji Sakurai; Masaki Ishigai; Toshiyuki Kudo; Kiyomi Ito
In a transcellular transport study, the apparent permeability coefficient (Papp) of a compound is evaluated using the range by which the amount of compound accumulated on the receiver side is assumed to be proportional to time. However, the time profile of the concentration of the compound in receiver (C3) often shows a lag time before reaching the linear range and later changes from linear to steady state. In this study, the linear range needed to calculate Papp in the C3-time profile was evaluated by a 3-compartment model. C3 was described by an equation with two steady states (C3=A3(1-e(-αt))+B3(1-e(-βt)), α>β), and by a simple approximate line (C3=A3-A3×αt) in the time range of 3/α<t<0.3/β; the lag time, defined as the interception of the x axis, was described as 1/α. The rate constant α was affected by the membrane permeability clearance and intracellular unbound fraction, while β was affected only by the former. The linear range that was approximated in the present study was not uniformly defined within the time interval in which C3 remains at <10% of the loading concentration, which is reported as the sink condition. In conclusion, this theoretical approach showed that the appropriate time range to evaluate Papp was 3/α<t<0.3/β.
Yakugaku Zasshi-journal of The Pharmaceutical Society of Japan | 2009
Takahiro Toda; Kanna Ohi; Toshiyuki Kudo; Tomoyuki Yoshida; Nobutomo Ikarashi; Kiyomi Ito; Kiyoshi Sugiyama
Biological & Pharmaceutical Bulletin | 2013
Junichi Iida; Toshiyuki Kudo; Kento Shimada; Yoshiyuki Yatsuno; Saori Yamagishi; Satoshi Hasegawa; Hideyuki Ike; Toru Sato; Hajime Kagaya; Kiyomi Ito
Journal of Pharmaceutical Sciences | 2017
Toshiyuki Kudo; Hitomi Goda; Yuki Yokosuka; Ryo Tanaka; Seina Komatsu; Kiyomi Ito
Drug Metabolism and Pharmacokinetics | 2017
Tomohisa Nakada; Toshiyuki Kudo; Toshiyuki Kume; Hiroyuki Kusuhara; Kiyomi Ito