Network


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

Hotspot


Dive into the research topics where Haruo Iwabuchi is active.

Publication


Featured researches published by Haruo Iwabuchi.


International Journal of Cancer | 1999

Antitumor activity and novel DNA-self-strand-breaking mechanism of CNDAC (1-(2-C-cyano-2-deoxy-?-d-ARABINO-Pentofuranosyl) cytosine) and itsN4-palmitoyl derivative (CS-682)

Kenji Hanaoka; Masako Suzuki; Tomowo Kobayashi; Fumie Tanzawa; Kazuo Tanaka; Takahiro Shibayama; Shinichi Miura; Tomoko Ikeda; Haruo Iwabuchi; Akihiko Nakagawa; Yoshihiro Mitsuhashi; Masashi Hisaoka; Masakatsu Kaneko; Akihiro Tomida; Yusuke Wataya; Tatsuji Nomura; Takuma Sasaki; Akira Matsuda; Takashi Tsuruo; Shinichi Kurakata

We have studied the antitumor activity and the novel DNA‐self‐strand‐breaking mechanism of CNDAC (1‐(2‐C‐cyano‐2‐deoxy‐β‐d‐arabino‐pentofuranosyl)cytosine) and its N4‐palmitoyl derivative (CS‐682). In vitro, CS‐682 showed strong cytotoxicity against human tumor cells comparable with that of CNDAC; both compounds displayed a similar broad spectrum. In vivo, however, orally administered CS‐682 showed a more potent activity against human tumor xenografts than CNDAC, 5′‐deoxy‐5‐fluorouridine, 5‐fluorouracil and 2′,2′‐difluorodeoxycytidine. Moreover, CS‐682 was effective against various human organ tumor xenografts at a wide dose range and with low toxicity, and was effective against P388 leukemic cells resistant to mitomycin‐C, vincristine, 5‐fluorouracil or cisplatin in syngeneic mice. CNDAC, an active metabolite of CS‐682, had a prolonged plasma half‐life after repeated oral administrations of CS‐682 but not after oral administrations of CNDAC itself. This difference may partially explain the higher antitumor activity of CS‐682 relative to CNDAC. In both CNDAC‐ and CS‐682‐treated carcinoma cells, CNDAC 5′‐triphosphate (CNDACTP) was generated and incorporated into a DNA strand. High performance liquid chromatography (HPLC) and mass spectrometric analysis of the nucleosides prepared by digestion of the DNA from the CNDAC‐treated cells detected ddCNC (2′‐C‐cyano‐2′,3′‐didehydro‐2′,3′‐dideoxycytidine), which was shown to be generated only when the self‐strand‐breakage of CNDACTP‐incorporated DNA occurred. The cytotoxicity of CNDAC was completely abrogated by the addition of 2′‐deoxycytidine and was low against cells with decreased deoxycytidine kinase. Our results suggest that CNDAC is converted to CNDACMP by deoxycytidine kinase and that the resulting CNDACTP incorporated into a DNA strand as CNDACMP may induce DNA‐self‐strand‐breakage. This novel DNA‐self‐strand‐breaking mechanism may contribute to the potent antitumor activity of CS‐682. Int. J. Cancer 82:226–236, 1999.


Pharmaceutical Research | 2001

Inhibition of in vitro metabolism of simvastatin by itraconazole in humans and prediction of in vivo drug-drug interactions

Michi Ishigam; Minoru Uchiyama; Tomoko Kondo; Haruo Iwabuchi; Shin-ichi Inoue; Wataru Takasaki; Toshihiko Ikeda; Toru Komai; Kiyomi Ito; Yuichi Sugiyama

AbstractPurpose. To evaluate an interaction between simvastatin and itraconazole in in vitro studies and to attempt a quantitative prediction of in vivo interaction in humans. Methods. The inhibitory effect of itraconazole on simvastatin metabolism was evaluated using human liver microsomes and the Ki values were calculated for the unbound drug in the reaction mixture. A physiologically-based pharmacokinetic model was used to predict the maximum in vivo drug-drug interaction. Results. Itraconazole competitively inhibited the metabolism of simvastatin to M-1 and M-2 with Ki values in the nM range. The area under the curve (AUC) of simvastatin after concomitant dosing with itraconazole was predicted to increase ca. 84-101-fold compared with that without administration of itraconazole. Taking into consideration the fact that this method predicts the maximum interaction, this agrees well with the clinical observation of a 19-fold increase. A similar prediction, based on the Ki value without taking into account the drug adsorption to microsomes, led to an underevaluation of the interaction. Conclusions. It was demonstrated that the competitive inhibition of CYP3A4-mediated simvastatin metabolism by itraconazole is the main cause of the drug interaction and that a Ki value corrected for drug adsorption to microsomes is the key factor in quantitatively predicting the maximum in vivo drug interactions.


Toxicological Sciences | 2010

Ethylene Glycol Monomethyl Ether–Induced Toxicity Is Mediated through the Inhibition of Flavoprotein Dehydrogenase Enzyme Family

Makoto Takei; Yosuke Ando; Wataru Saitoh; Tomoe Tanimoto; Naoki Kiyosawa; Sunao Manabe; Atsushi Sanbuissho; Osamu Okazaki; Haruo Iwabuchi; Takashi Yamoto; Klaus-Peter Adam; James E. Weiel; John A. Ryals; Michael V. Milburn; Lining Guo

Ethylene glycol monomethyl ether (EGME) is a widely used industrial solvent known to cause adverse effects to human and other mammals. Organs with high metabolism and rapid cell division, such as testes, are especially sensitive to its actions. In order to gain mechanistic understanding of EGME-induced toxicity, an untargeted metabolomic analysis was performed in rats. Male rats were administrated with EGME at 30 and 100 mg/kg/day. At days 1, 4, and 14, serum, urine, liver, and testes were collected for analysis. Testicular injury was observed at day 14 of the 100 mg/kg/day group only. Nearly 1900 metabolites across the four matrices were profiled using liquid chromatography-mass spectrometry/mass spectrometry and gas chromatography-mass spectrometry. Statistical analysis indicated that the most significant metabolic perturbations initiated from the early time points by EGME were the inhibition of choline oxidation, branched-chain amino acid catabolism, and fatty acid β-oxidation pathways, leading to the accumulation of sarcosine, dimethylglycine, and various carnitine- and glycine-conjugated metabolites. Pathway mapping of these altered metabolites revealed that all the disrupted steps were catalyzed by enzymes in the primary flavoprotein dehydrogenase family, suggesting that inhibition of flavoprotein dehydrogenase–catalyzed reactions may represent the mode of action for EGME-induced toxicity. Similar urinary and serum metabolite signatures are known to be the hallmarks of multiple acyl-coenzyme A dehydrogenase deficiency in humans, a genetic disorder because of defects in primary flavoprotein dehydrogenase reactions. We postulate that disruption of key biochemical pathways utilizing flavoprotein dehydrogenases in conjugation with downstream metabolic perturbations collectively result in the EGME-induced tissue damage.


Drug Metabolism and Disposition | 2006

Characterization of phenotypes in Gstm1-null mice by cytosolic and in vivo metabolic studies using 1,2-dichloro-4-nitrobenzene.

Kazunori Fujimoto; Shingo Arakawa; Yukari Shibaya; Hiroaki Miida; Yosuke Ando; Hiroaki Yasumo; Ayako Hara; Minoru Uchiyama; Haruo Iwabuchi; Wataru Takasaki; Sunao Manabe; Takashi Yamoto

Glutathione S-transferase Mu 1 (GSTM1) has been regarded as one of the key enzymes involved in phase II reactions in the liver, because of its high expression level. In this study, we generated mice with disrupted glutathione S-transferase Mu 1 gene (Gstm1-null mice) by gene targeting, and characterized the phenotypes by cytosolic and in vivo studies. The resulting Gstm1-null mice appeared to be normal and were fertile. Expression analyses for the Gstm1-null mice revealed a deletion of Gstm1 mRNA and a small decrease in glutathione S-transferase alpha 3 mRNA. In the enzymatic study, GST activities toward 1,2-dichloro-4-nitrobenzene (DCNB) and 1-chloro-2,4-dinitrobenzene (CDNB) in the liver and kidney cytosols were markedly lower in Gstm1-null mice than in the wild-type control. Gstm1-null mice had GST activities of only 6.1 to 21.0% of the wild-type control to DCNB and 26.0 to 78.6% of the wild-type control to CDNB. After a single oral administration of DCNB to Gstm1-null mice, the plasma concentration of DCNB showed larger AUC0–24 (5.1–5.3 times, versus the wild-type control) and higher Cmax (2.1–2.2 times, versus the wild-type control), with a correspondingly lower level of glutathione-related metabolite (AUC0–24, 9.4–17.9%; and Cmax, 9.7–15.6% of the wild-type control). In conclusion, Gstm1-null mice showed markedly low ability for glutathione conjugation to DCNB in the cytosol and in vivo and would be useful as a deficient model of GSTM1 for absorption, distribution, metabolism, and excretion/toxicology studies.


Drug Metabolism and Disposition | 2010

Biotransformation of prasugrel, a novel thienopyridine antiplatelet agent, to the pharmacologically active metabolite.

Katsunobu Hagihara; Miho Kazui; Atsushi Kurihara; Haruo Iwabuchi; Minoru Ishikawa; Hiroyuki Kobayashi; Naoki Tanaka; Osamu Okazaki; Nagy A. Farid; Toshihiko Ikeda

Prasugrel, a novel thienopyridine antiplatelet agent, undergoes rapid hydrolysis in vivo to a thiolactone, R-95913, which is further converted to its thiol-containing, pharmacologically active metabolite, R-138727, by oxidation via cytochromes P450 (P450). We trapped a sulfenic acid metabolite as a mixed disulfide with 2-nitro-5-thiobenzoic acid in an incubation mixture containing the thiolactone R-95913, expressed CYP3A4, and NADPH. Further experiments investigated one possible mechanism for the conversion of the sulfenic acid to the active thiol metabolite in vitro. A mixed disulfide form of R-138727 with glutathione was found to be a possible precursor of R-138727 in vitro when glutathione was present. The rate constant for the reduction of the glutathione conjugate of R-138727 to R-138727 was increased by addition of human liver cytosol to the human liver microsomes. Thus, one possible mechanism for the ultimate formation of R-138727 in vitro can be through formation of a sulfenic acid mediated by P450s followed possibly by a glutathione conjugation to a mixed disulfide and reduction of the disulfide to the active metabolite R-138727.


Drug Metabolism and Disposition | 2006

HUMAN UDP-GLUCURONOSYLTRANSFERASE, UGT1A8, GLUCURONIDATES DIHYDROTESTOSTERONE TO A MONOGLUCURONIDE AND FURTHER TO A STRUCTURALLY NOVEL DIGLUCURONIDE

Takahiro Murai; Naozumi Samata; Haruo Iwabuchi; Toshihiko Ikeda

We identified human UDP-glucuronosyltransferase (UGT) isoforms responsible for producing dihydrotestosterone (DHT) diglucuronide, a novel glucuronide in which the second glucuronosyl moiety is attached at the C2′ position of the first glucuronosyl moiety, leading to diglucuronosyl conjugation of a single hydroxyl group of DHT at the C17 position. Incubation of the DHT monoglucuronide with 12 cDNA-expressed recombinant human UGT isoforms and uridine 5′-diphosphoglucuronic acid resulted in a low but measurable DHT diglucuronidation activity primarily with UGT1A8, a gastrointestinal UGT, and to a lesser extent with UGT1A1 and UGT1A9. In contrast, the activity of DHT monoglucuronidation was high and was found in UGT2B17, UGT2B15, UGT1A8, and UGT1A4 in descending order. Among the 12 UGT isoforms tested, only UGT1A8 was capable of producing DHT diglucuronide from DHT. The kinetics of DHT diglucuronidation by microsomes from human liver and intestine fitted the Michaelis-Menten model, and the Vmax/Km value for the intestinal microsomes was approximately 4 times greater than that for the liver microsomes.


Drug Metabolism and Disposition | 2010

Identification of novel metabolic pathways of pioglitazone in hepatocytes: N-glucuronidation of thiazolidinedione ring and sequential ring opening pathway

Minoru Uchiyama; Thomas Fischer; Juergen Mueller; Minoru Oguchi; Naotoshi Yamamura; Hiroko Koda; Haruo Iwabuchi; Takashi Izumi

The metabolism of [14C]pioglitazone was studied in vitro in incubations with freshly isolated human, rat, and monkey hepatocytes. Radioactivity detection high-performance liquid chromatography analysis of incubation extracts showed the detection of 13 metabolites (M1–M13) formed in incubations with human hepatocytes. An identical set of metabolites (M1–M13) was also detected in monkey hepatocytes. However, in rat hepatocytes, M1 through M3, M5 through M7, M9 through M11, and M13 were also detected, but M4, M8, and M12 were not detected. The structures of the metabolites were elucidated by liquid chromatography/tandem mass spectrometry using electrospray ionization. Novel metabolites of pioglitazone detected using these methods included thiazolidinedione ring-opened methyl sulfoxide amide (M1), thiazolidinedione ring-opened N-glucuronide (M2), thiazolidinedione ring-opened methyl sulfone amide (M3), thiazolidinedione ring N-glucuronide (M7), thiazolidinedione ring-opened methylmercapto amide (M8), and thiazolidinedione ring-opened methylmercapto carboxylic acid (M11). In summary, based on the results from these studies, two novel metabolic pathways for pioglitazone in hepatocytes are proposed to be as follows: 1) N-glucuronidation of the thiazolidinedione ring of pioglitazone to form M7 followed by hydrolysis to M2, and methylation of the mercapto group of the thiazolidinedione ring-opened mercapto carboxylic acid to form M11; and 2) methylation of the mercapto group of the thiazolidinedione ring-opened mercapto amide to form M8, oxidation of M8 to form M1, and oxidation of M1 to form M3.


Xenobiotica | 1988

Induction of Peroxisome Proliferation in Rat Liver by Dietary Treatment with 2,2,4,4,6,8,8-Heptamethylnonane

Toshihiko Ikeda; M. Ida-Enomoto; I. Mori; K. Fukuda; Haruo Iwabuchi; T. Komai; Tetsuya Suga

1. Exposure of rats to 1% (w/w) of 2,2,4,4,6,8,8-heptamethylnonane in the diet for 2 weeks resulted in marked induction of liver peroxisome proliferation as judged from electron micrography, elevated activities of hepatic catalase (36%), cyanide-insensitive palmitoyl-CoA oxidase (10-fold), carnitine acetyl transferase (9.6-fold), lauric acid hydroxylase (12.4-fold), and the induction of the 80 K protein in SDS-polyacrylamide gel electrophoresis (4.1-fold). 2. 2,2,4,4,6,8,8-Heptamethylnonane dicarboxylic acid, a non-beta-oxidizable fatty acid, was detected as the major metabolite in the liver, an example of an unmetabolizable lipophilic anion as a peroxisome proliferator.


Drug Metabolism and Disposition | 2011

Pharmacokinetics, metabolism, and disposition of rivoglitazone, a novel peroxisome proliferator-activated receptor γ agonist, in rats and monkeys

Minoru Uchiyama; Haruo Iwabuchi; Fujiko Tsuruta; Koji Abe; Makoto Takahashi; Hiroko Koda; Minoru Oguchi; Osamu Okazaki; Takashi Izumi

The pharmacokinetics, metabolism, and excretion of rivoglitazone [(RS)-5-{4-[(6-methoxy-1-methyl-1H-benzimidazol-2-yl)methoxy]benzyl}-1,3-thiazolidine-2,4-dione monohydrochloride], a novel thiazolidinedione (TZD) peroxisome proliferator-activated receptor γ selective agonist, were evaluated in male F344/DuCrlCrlj rats and cynomolgus monkeys. The total body clearance and volume of distribution of rivoglitazone were low in both animals (0.329–0.333 ml per min/kg and 0.125–0.131 l/kg for rats and 0.310–0.371 ml per min/kg and 0.138–0.166 l/kg for monkeys), and the plasma half-life was 4.55 to 4.84 h for rats and 6.21 to 6.79 h for monkeys. The oral bioavailability was high (>95% in rats and >76.1% in monkeys), and the exposure increased dose proportionally. After administration of [14C]rivoglitazone, radioactivity was mainly excreted in feces in rats, whereas radioactivity was excreted in urine and feces with the same ratio in monkeys. Because excreted rivoglitazone in urine and bile was low, metabolism was predicted to be the main contributor to total body clearance. The structures of 20 metabolites (M1–M20) were identified, and 5 initial metabolic pathways were proposed: O-demethylation, TZD ring opening, N-glucuronidation, N-demethylation, and TZD ring hydroxylation. O-Demethylation was the main metabolic pathway in both animals, but N-demethylation and TZD ring hydroxylation were observed only in monkeys. N-Glucuronide (M13) was nonenzymatically hydrolyzed to TZD ring-opened N-glucuronide (M9), and the amount of these metabolites in monkeys was larger than that in rats. In plasma, rivolitazone was observed as the main component in both animals, and O-demethyl-O-sulfate (M11) was observed as the major metabolite in rats but as many minor metabolites in monkeys.


Journal of Pharmaceutical and Biomedical Analysis | 2012

Glutathione S-transferase pi trapping method for generation and characterization of drug–protein adducts in human liver microsomes using liquid chromatography–tandem mass spectrometry

Hideo Yukinaga; Haruo Iwabuchi; Osamu Okazaki; Takashi Izumi

Covalent binding of reactive metabolites (RMs) to proteins is thought to play an important role in the processes leading to adverse drug reactions. Therefore, there is great interest in methodologies that enable the characterization of covalent binding of drugs to proteins. To facilitate the study of drug-protein adducts, we have developed a liquid chromatography-tandem mass spectrometry (LC-MS/MS) method for characterizing RM-modified proteins formed through drug bioactivation in human liver microsomes (HLMs), which are commonly used for the in vitro drug bioactivation studies. The technique was illustrated by the trapping of RMs of acetaminophen (APAP) and raloxifene with human glutathione S-transferase pi (hGSTP) as a model target protein. After hGSTP-supplemented HLM incubations, the modified/unmodified hGSTP fractions were collected by high-performance liquid chromatography. hGSTP fractions were digested with trypsin, and then analyzed by linear ion trap-orbitrap mass spectrometry followed by a SEQUEST database search. Characteristic MS/MS fragment ions of RM-modified peptides were identified by searching for possible adducted-mass shifts. The method successfully revealed that RMs of both drugs adducted to Cys-47 of hGSTP and the mass shifts corresponded to modification by the N-acetyl-p-benzoquinone imine form of APAP and diquinone methide form of raloxifene, respectively. The developed method would be a possible tool for widespread use for the generation and characterization of drug-protein adducts in HLMs and has the potential to assess the risk of covalent binding of drugs to proteins.

Collaboration


Dive into the Haruo Iwabuchi's collaboration.

Top Co-Authors

Avatar

Toshihiko Ikeda

Yokohama College of Pharmacy

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hiroshi Kogen

Meiji Pharmaceutical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge