Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Asako Nishimura is active.

Publication


Featured researches published by Asako Nishimura.


European Journal of Pharmaceutical Sciences | 2012

Controlled release of insulin from self-assembling nanofiber hydrogel, PuraMatrix™: application for the subcutaneous injection in rats.

Asako Nishimura; Taro Hayakawa; Yu Yamamoto; Mami Hamori; Keiko Tabata; Keiko Seto; Nobuhito Shibata

The concept of this research is, using the acetyl-(Arg-Ala-Asp-Ala)₄-CONH₂ peptide hydrosol (PuraMatrix™, PM), to develop an new injectable formula of controlled insulin delivery for subcutaneous injection. PM has sol-gel phase transition behavior, and was developed as a scaffold in the field of tissue engineering. The aqueous media of the PM including insulin changed from a sol to a gel phase with increasing ion strength of phosphate ion and pH in working environments in vitro and in vivo. In this study, we examined the in vitro insulin dissolution behavior and the in vivo pharmacokinetics and pharmacodynamics after subcutaneous administration of PM-insulin sol (PM-Isol). In the in vitro release study, after PM-Isol was converted to a gel phase (PM-Igel), PM concentration-dependent and controlled release of insulin were observed at the final concentrations of PM between 0.1% and 2.0% (w/v). The PM-Isol is changed to gel form in vivo, and exhibited a sustained-release pharmacokinetics of insulin, where PM concentration-dependent prolongation of efficacy was found. The plasma glucose level markedly decreased, and the lowest plasma glucose level was maintained up to 24h when 2.0% (w/v) PM-Isol was administered subcutaneously to rats. The PM-Isol, we developed here, is applicable for the wild-type of insulin, and increased the bioavailability and hypoglycemic efficacy of insulin after subcutaneous injection. Hence, the PM is a useful inactive ingredient to produce various types of control-released system of insulin by making just a few changes in PM content of the formulation.


International Journal of Pharmaceutics | 2014

Preparation and pharmaceutical evaluation of nano-fiber matrix supported drug delivery system using the solvent-based electrospinning method.

Mami Hamori; Shiori Yoshimatsu; Yuki Hukuchi; Yuki Shimizu; Keizo Fukushima; Nobuyuki Sugioka; Asako Nishimura; Nobuhito Shibata

In this study, utilizing the solvent-based electrospinning (ES) method, which is mainly employed in the textile industry, we prepared nanofiber-based capsules including drugs for controlled-release delivery systems using methacrylic acid copolymer (EUDRAGIT(®) S100, MAC) as a polymer, and evaluated their in vitro drug dissolution profiles and in vivo pharmacokinetics in rats. As the model drugs, uranine (UN) was used as a water-soluble drug and nifedipine (NP) as a water-insoluble drug. The mean diameters of drug free nano-fiber and nano-fiber including NP or UN were 751.5 ± 67.2, 703.3 ± 71.2 and 2477.8 ± 206.1 nm, respectively. X-ray diffraction for the nano-fibrotic sheet showed that UN and/or NP were packed in nano-fiber in an amorphous form. The in vitro release of UN or NP from the nano-fiber packed capsules (NFPC) and milled-powder of nano-fiber packed capsules (MPPC) showed controlled release of UN or NP as compared to capsules of a physical mixture of MAC and each drug. An in vivo pharmacokinetic study in rats after intraduodenal administration of NFPC or MPPC including UN and/or NP clearly demonstrated that application of nano-fibrotic technique as a drug delivery system offers drastic changes in pharmacokinetic profiles for both water-soluble and water-insoluble drugs. The ES method is a useful technique to prepare a nano-fiber like solid dispersion for polar or nonpolar drugs, and has wide potential pharmaceutical applications.


Biomedicine & Pharmacotherapy | 2009

Effect of cyclosporine on drug transport and pharmacokinetics of nifedipine.

Madhura Dorababu; Asako Nishimura; Thangavelu Prabha; Kazumasa Naruhashi; Nobuyuki Sugioka; Kanji Takada; Nobuhito Shibata

Nifedipine (NFP) is an anti-hypersensitive drug and a well-known substrate of cytochrome P450 3A4 (CYP3A4), while cyclosporine (CSP) is a potent p-glycoprotein (P-gp) inhibitor. P-gp is a drug transporter, which determines the absorption and bioavailability of many drugs that are substrates for P-gp. Drugs that induce or inhibit P-gp may have a profound effect on the absorption and pharmacokinetics (PK) of drugs transported by P-gp within the body, possibly compromising their bioavailability. But the role of P-gp in the NFP efflux and its impact on PK profile is not known. Hence in our present study we attempted to investigate the effect of CSP on oral absorption and PK of NFP. Rhodamine 123 (Rho 123), a known P-gp substrate was used as a positive control. Male Wistar rats (350-400 g) were used for the study. Rats were divided into 4 groups (n=6 each); one group was treated with vehicle (cremophor) followed by NFP (0.2 mg/kg; i.v. bolus) and the other group with CSP (10 mg/kg; i.v.) followed by NFP. Group 3 and 4 were treated with vehicle (cremophor) followed by Rho 123 (0.2 mg/kg, i.v.) and CSP (10 mg/kg; i.v.) followed by Rho 123 (0.2 mg/kg, i.v.) respectively. The blood samples were collected at 0, 5, 10, 15, 30, 60, 90, 120, 180 and 240 min after NFP administration. NFP concentrations in plasma were analyzed by LC-MS-MS and Rho 123 was analyzed by fluorimetric detector. NFP efflux was significantly decreased in CSP treated rats (49.1% decrease, P<0.05), while NFP concentration in plasma were not changed. However the decrease in NFP efflux did not show any significant changes in NFP PK parameters (T(max); 2.0 vs. 2.5 min, C(max); 0.084 vs. 0.076 microg/ml, T(1/2); 84.0 vs. 91.4 min, AUC(0-t); 4.183 vs. 3.467 microg h/ml, AUC(infinity); 5.915 vs. 4.769 microg h/ml, AUMC(0-t); 224.073 vs. 173.063 microg h/ml, AUMC(infinity); 776.871 vs. 575.038 microg h/ml, MRT(0-t); 53.608 vs. 49.538 microg h/ml, MRT(infinity); 118.194 vs. 115.246 microg h/ml, CL(tot); 0.0375 vs. 0.0433 l/h, Vd(ss); 3.999 vs. 4.641 l in NFP alone vs. CSP+NFP groups respectively). Thus the results indicate that NFP would belong to a group of P-gp substrate. The decrease in efflux of NFP by CSP, through inhibition of P-gp, into the intestinal lumen did not show any impact on PK. This could be due to the activity of other transporters and/or CYP3A4 may have more limiting role than P-gp on NFP metabolism and disposition that is why inhibiting P-gp did not lead to increase the bioavailability and PK alterations.


Xenobiotica | 2014

Semi-physiological pharmacokinetic–pharmacodynamic modeling and simulation of 5-fluorouracil for the whole time course of alterations in leukocyte, neutrophil and lymphocyte counts in rats

Shinji Kobuchi; Yukako Ito; Taro Hayakawa; Shota Kuwano; Akiko Baba; Kota Shinohara; Asako Nishimura; Nobuhito Shibata; Kanji Takada

Abstract 1. We aimed to develop a simple pharmacokinetic–pharmacodynamic (PK–PD) model to predict the onset and degree of severe toxic side effects that severely limit the use of many anticancer agents, such as myelosuppression, in rats. 2. Our PK–PD model consisted of a two-compartment PK model, with one compartment representing proliferative cells and some transit compartments consisting of maturing cells, while the other compartment represented circulating blood cells for the PD model. 3. The semi-physiological PK–PD model effectively captured the features of myelosuppression and the degree of the off-target toxicities observed after 5-fluorouracil (5-FU) chemotherapy, and helped simultaneously simulate the whole time course for alterations in leukocyte, neutrophil and lymphocyte counts after 5-FU treatment in rats. Interestingly, by plotting the nadir period of leukocyte, neutrophil and lymphocyte counts as determined by PK–PD analytical simulation curves against the area under the plasma 5-FU concentration–time curve (AUC0–∞) after intravenous administration of 5-FU, a linear relationship was inferred, with r2 = 0.989, 0.877 and 0.956, respectively. 4. The semi-physiological PK–PD model is a valuable tool for evaluating a variety of novel cancer chemopreventive agents or emerging therapeutic strategies that are difficult to address in humans.


Biopharmaceutics & Drug Disposition | 2013

A predictive biomarker for altered 5-fluorouracil pharmacokinetics following repeated administration in a rat model of colorectal cancer

Shinji Kobuchi; Shota Kuwano; Kazuki Imoto; Kae Okada; Asako Nishimura; Yukako Ito; Nobuhito Shibata; Kanji Takada

The relationship between the plasma ratio of dihydrouracil/uracil (UH2/Ura) and hepatic dihydropyrimidine dehydrogenase (DPD) activity after repeated 5‐fluorouracil (5‐FU) treatment in rats with colorectal cancer (CRC) was investigated. Repeated intravenous 5‐FU bolus injections resulted in a significant decrease in the total clearance (CLtot) and an increased area under the curve (AUC0‐∞) in CRC rats. Furthermore, the hepatic DPD levels and the plasma ratio of UH2/Ura decreased significantly and lost their circadian rhythms in CRC rats treated repeatedly with 5‐FU, although significant circadian variation in the two parameters was observed in the control CRC rats. Moreover, a significant correlation was found between the plasma ratio of UH2/Ura and hepatic DPD activity in CRC rats untreated and treated with single or repeated 5‐FU administration (r2 = 0.865, p < 0.01). The ratio of UH2/Ura in plasma could be a predictive biomarker of the suppression of hepatic DPD levels during repeated 5‐FU‐based treatment. Furthermore, by plotting the observed pharmacokinetic parameters of 5‐FU against hepatic DPD activity levels predicted by the ratio of UH2/Ura in plasma, AUC0‐∞, CLtot and half‐life (t1/2) were closely linked to predicted hepatic DPD activity levels. These observations suggest that the factor that significantly influences the AUC0‐∞, CLtot and t1/2 of 5‐FU after single or repeated administration of 5‐FU is the hepatic DPD activity and it could be assessed by the ratio of UH2/Ura in plasma. Copyright


Journal of Pharmaceutical Sciences | 2016

Population Pharmacokinetic–Toxicodynamic Modeling and Simulation of Cisplatin-Induced Acute Renal Injury in Rats: Effect of Dosing Rate on Nephrotoxicity

Keizo Fukushima; Akira Okada; Kosho Sasaki; Shuichi Kishimoto; Shoji Fukushima; Mai Hamori; Asako Nishimura; Nobuhito Shibata; Toshiharu Shirai; Ryu Terauchi; Toshikazu Kubo; Nobuyuki Sugioka

Nephrotoxicity is the major dose-limiting toxicity of cisplatin (CDDP). The aim of this study was to develop a pharmacokinetic (PK)/toxicodynamic (TD) model of CDDP-induced acute renal injury in rats and to simulate nephrotoxicity at various dosing rates. CDDP was administered to rats by a 30-s bolus or a 2-h infusion (1.0, 2.5, 5.0, and 7.5 mg/kg). Unbound CDDP concentrations in plasma and urine were determined up to 2 h after administration in the PK study, and plasma creatinine (Cr) levels were monitored for up to 7 days as an index of nephrotoxicity in the TD study. The PK was linear and was fitted with a traditional 2-compartment model. The TD was nonlinear and differed between dosing rates. The creatinine concentration profiles were fitted with a signal transduction-indirect response model. Population analysis using a nonlinear mixed-effect model was adapted to the developed PK/TD model and was well-validated. Dosing simulations from the developed population PK/TD model indicated that CDDP-induced nephrotoxicity was due to not only Cmax but also the time above the toxic concentration of CDDP. Prolongation of infusion time will not necessarily attenuate acute nephrotoxicity. This study demonstrated the potential utility of PK/TD modeling for preventing nephrotoxicity.


Biomedicine & Pharmacotherapy | 2016

Preparation and pharmaceutical evaluation of acetaminophen nano-fiber tablets: Application of a solvent-based electrospinning method for tableting

Mami Hamori; Kana Nagano; Sayaka Kakimoto; Kazumasa Naruhashi; Akiko Kiriyama; Asako Nishimura; Nobuhito Shibata

In this study, we developed nano-fiber-based tablets with acetaminophen (AAP; LogPow=0.51) for controlled-release delivery systems and evaluated in vitro drug dissolution and in vivo pharmacokinetics in rats. Nano-fibers made from methacrylic acid copolymer S (MAC; EUDRAGIT S100) and containing AAP were prepared using a solvent-based electrospinning (ES) method. In vitro dissolution rate profiles of AAP showed tableting pressure-dependent decreases and pH-dependent increases. The results of tablet tracking by X-ray irradiation showed tablets based on MAC nano-fibers did not disintegrate in the upper intestinal lumen and had the properties of a long-term-acting tablet. In addition, the in vitro release profiles of AAP from nano-fiber tablets prepared by dissolving MAC with AAP (NFT), nano-fiber tablets prepared by adsorbing AAP to drug-free MAC nano-fibers (NFTadso), and tablets prepared by adsorbing half the amount of AAP to MAC nano-fibers containing the remaining amount of AAP (NFThalf) showed independent controlled-release aspects of AAP compared with physical mixture tablets (PMT). In vivo pharmacokinetic studies in rats after intraduodenal administration of 14 mg/rat AAP in NFT, NFTadso, and NFThalf demonstrated that all these tablets based on MAC nano-fibers showed sustained-release profiles compared with PMT, and showed ultra-sustained release properties for AAP. These new tablets based on MAC nano-fibers did not disintegrate in the intestine in the lower pH region, and the tablets could regulate the release of AAP in a pH-dependent manner. The ES method is a useful technique to prepare nano-fibers and showed promising results as an oral delivery system for sustained-release regulation.


Xenobiotica | 2015

Semi-physiological pharmacokinetic–pharmacodynamic (PK–PD) modeling and simulation of 5-fluorouracil for thrombocytopenia in rats

Shinji Kobuchi; Yukako Ito; Taro Hayakawa; Asako Nishimura; Nobuhito Shibata; Kanji Takada; Toshiyuki Sakaeda

Abstract 1. The aim of this study was to develop a simple pharmacokinetic–pharmacodynamic (PK–PD) model that could characterize the complete time-course of alterations in platelet counts to predict the onset and degree of thrombocytopenia, which severely limits the use of the anticancer agent 5-fluorouracil (5-FU), in rats. 2. Platelet counts were measured in rats following the intravenous administration of various doses of 5-FU for 4 days to obtain data for an analysis of the PK–PD model. Our PK–PD model consisted of a two-compartment PK model, with three compartments for the PD model and 10 structural PK–PD model parameters. 3. After the 5-FU treatment, platelet counts transiently decreased to a nadir level, showed a rebound to above the baseline level before recovering to baseline levels. Nadir platelet counts and rebounds varied with the AUC0–∞ level. The final PK–PD model effectively characterized platelet count data and final PD parameters were estimated with high certainty. 4. This PK–PD model and simulation may represent a valuable tool for quantifying and predicting the complete time-course of alterations in blood cell counts, and could contribute to the development of therapeutic strategies with 5-FU and assessments of various novel anticancer agents that are difficult to examine in humans.


Journal of Pharmacological and Toxicological Methods | 2014

Pharmacokinetic–pharmacodynamic (PK–PD) modeling and simulation of 5-fluorouracil for erythropenia in rats

Shinji Kobuchi; Yukako Ito; Taro Hayakawa; Asako Nishimura; Nobuhito Shibata; Kanji Takada; Toshiyuki Sakaeda

INTRODUCTION The aim of the present study was to develop a simple pharmacokinetic-pharmacodynamic (PK-PD) model in rats that could predict the onset and degree of erythropenia, a severely toxic side effect that severely limits the use of the anticancer agent 5-fluorouracil (5-FU). METHODS Total erythrocyte counts, hemoglobin (Hb) concentrations, and hematocrit (Hct) levels were measured in rats following the intravenous bolus administration of 5-FU for 4 days in order to obtain data for an analysis of the PK-PD model. Our PK-PD model consisted of a two-compartment PK model, with two compartments for the PD model and nine structural PK-PD model parameters. RESULTS After the intravenous bolus administration of 5, 10, or 20 mg/kg of 5-FU to rats, absolute erythrocyte counts, Hb concentrations, and Hct levels transiently decreased, reached minimum levels on Days 7-14, and then returned to baseline levels. The nadir values (Cnadir) for rats treated with 5, 10, or 20 mg/kg of 5-FU were significantly decreased to approximately 79.4, 76.3, or 46.5% of the baseline value (Cbaseline) in erythrocyte counts, 86.3, 83.3, or 45.7% of Cbaseline in Hb concentrations, 88.6, 85.5, or 47.1% of Cbaseline in Hct levels, respectively. The PK-PD model effectively captured the features of erythropenia and Cnadir after 5-FU chemotherapy. This PK-PD model was successfully used to characterize the learner relationship between the area under the plasma 5-FU concentration-time curve (AUC0-∞) following the intravenous bolus administration of 5-FU and the Cnadir in erythrocyte counts, Hb concentrations, and Hct levels after the 5-FU treatment. DISCUSSION The results of the present study suggest that the administration of a pharmacokinetically modified dose of 5-FU could minimize the Cnadir in erythrocyte counts, Hb concentrations, and Hct levels following the administration of 5-FU. The PK-PD model and simulation represent valuable approaches for quantifying and predicting erythropenia as well as determining individual doses and the time at which the subsequent course of the treatment should start.


Biological & Pharmaceutical Bulletin | 2017

Alterations in Cisplatin Pharmacokinetics and Its Acute/Sub-chronic Kidney Injury over Multiple Cycles of Cisplatin Treatment in Rats

Akira Okada; Keizo Fukushima; Mai Fujita; Mana Nakanishi; Mami Hamori; Asako Nishimura; Nobuhito Shibata; Nobuyuki Sugioka

Cisplatin (CDDP)-induced acute kidney injury (AKI) is a major clinical concern. CDDP treatment is generally conducted with multiple cycles; the magnitude of the CDDP-induced AKI may be altered by these cycles. Moreover, sub-chronic kidney injury (sCKI) induced by repeated CDDP treatment is often associated with renal interstitial fibrosis, potentially leading to chronic kidney disease. Therefore, it is suggested that the management of not only AKI but also sCKI induced by CDDP in multiple cycles plays an important role in the outcome of CDDP-based chemotherapy. This study investigated the alteration in pharmacokinetics and toxicodynamics of CDDP that was repeatedly administered for three cycles in rats; a cycle consisted of CDDP (5.0 mg/kg, bolus injection) followed by a 21-d washout period. AKI and sCKI were evaluated by plasma creatinine concentration. In repeated multiple administration of CDDP, renal clearance was decreased and the amounts of accumulated Pt in kidneys increased by the cycle. AKI and sCKI were similarly exacerbated by the cycle, whereas the degree of AKI showed a large inter- and intra-individual variation in each cycle. However, the degree of sCKI constantly increased (creatinine increasing ratio in any cycle is about 150%), suggesting that the degree of sCKI in any given cycle was predictable by monitoring the initial creatinine baseline. In this study, therefore, it is suggested that the evaluation of sCKI by monitoring creatinine concentration at base is important for the estimation of CDDP-induced nephrotoxicity. These results may provide useful information for more effective and safe CDDP-based chemotherapy with evidence-based dose adjustment.

Collaboration


Dive into the Asako Nishimura's collaboration.

Top Co-Authors

Avatar

Nobuhito Shibata

Doshisha Women's College of Liberal Arts

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kanji Takada

Kyoto Pharmaceutical University

View shared research outputs
Top Co-Authors

Avatar

Mami Hamori

Doshisha Women's College of Liberal Arts

View shared research outputs
Top Co-Authors

Avatar

Akira Okada

Kobe Gakuin University

View shared research outputs
Top Co-Authors

Avatar

Kazumasa Naruhashi

Doshisha Women's College of Liberal Arts

View shared research outputs
Top Co-Authors

Avatar

Taro Hayakawa

Shiga University of Medical Science

View shared research outputs
Top Co-Authors

Avatar

Yukako Ito

Kyoto Pharmaceutical University

View shared research outputs
Top Co-Authors

Avatar

Shinji Kobuchi

Kyoto Pharmaceutical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge