Network


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

Hotspot


Dive into the research topics where Kenji Furutsuka is active.

Publication


Featured researches published by Kenji Furutsuka.


Nuclear Medicine and Biology | 2003

[11C]DAA1106: radiosynthesis and in vivo binding to peripheral benzodiazepine receptors in mouse brain

Ming-Rong Zhang; Takayo Kida; Junko Noguchi; Kenji Furutsuka; Jun Maeda; Tetsuya Suhara; Kazutoshi Suzuki

DAA1106 (N-(2,5-Dimethoxybenzyl)-N-(5-fluoro-2-phenoxyphenyl)acetamide), is a potent and selective ligand for peripheral benzodiazepine receptors (PBR) in mitochondrial fractions of rat (K(i)=0.043 nM) and monkey (K(i)=0.188 nM) brains. This compound was labeled by [(11)C]methylation of a corresponding desmethyl precursor (DAA1123) with [(11)C]CH(3)I in the presence of NaH, with a 72+/-16% (corrected for decay) incorporation yield of radioactivity. After HPLC purification, [(11)C]DAA1106 was obtained with > or =98% radiochemical purity and specific activity of 90-156 GBq/micromol at the end of synthesis. After iv injection of [(11)C]DAA1106 into mice, high accumulations of radioactivity were found in the olfactory bulb and cerebellum, the high PBR density regions in the brain. Coinjection of [(11)C]DAA1106 with unlabeled DAA1106 and PBR-selective PK11195 displayed a significant reduction of radioactivity, suggesting a high specific binding of [(11)C]DAA1106 to PBR. Although this tracer was rapidly metabolized in the plasma, only [(11)C]DAA1106 was detected in the brain tissues, suggesting the specific binding in the brain due to the tracer itself. These findings revealed that [(11)C]DAA1106 is a potential and selective positron emitting radioligand for PBR.


Bioorganic & Medicinal Chemistry Letters | 2003

[18F]FMDAA1106 and [18f]FEDAA1106: Two positron-emitter labeled ligands for peripheral benzodiazepine receptor (PBR)

Ming-Rong Zhang; Jun Maeda; Kenji Furutsuka; Yuichiro Yoshida; Masanao Ogawa; Tetsuya Suhara; Kazutoshi Suzuki

We synthesized and evaluated N-(5-fluoro-2-phenoxyphenyl)-N-(2-[(18)F]fluoromethyl-5-methoxybenzyl)acetamide ([(18)F]-FMDAA1106) and N-(5-fluoro-2-phenoxyphenyl)-N-(2-[(18)F]fluoroethyl-5-methoxybenzyl)acetamide ([(18)F]FEDAA1106) as two potent radioligands for peripheral benzodiazepine receptors (PBR). [(18)F]FMDAA1106 and [(18)F]FEDAA1106 were respectively synthesized by fluoroalkylation of the desmethyl precursor DAA1123 with [(18)F]FCH(2)I and [(18)F]FCH(2)CH(2)Br. Ex vivo autoradiograms of [(18)F]FMDAA1106 and [(18)F]FEDAA1106 binding sites in the rat brains revealed that a high radioactivity was present in the olfactory bulb, the highest PBR density region in the brain.


The Journal of Nuclear Medicine | 2014

Radiosynthesis, Photoisomerization, Biodistribution, and Metabolite Analysis of 11C-PBB3 as a Clinically Useful PET Probe for Imaging of Tau Pathology

Hiroki Hashimoto; Kazunori Kawamura; Nobuyuki Igarashi; Makoto Takei; Tomoya Fujishiro; Yoshiharu Aihara; Satoshi Shiomi; Masatoshi Muto; Takehito Ito; Kenji Furutsuka; Tomoteru Yamasaki; Joji Yui; Lin Xie; Maiko Ono; Akiko Hatori; Kazuyoshi Nemoto; Tetsuya Suhara; Makoto Higuchi; Zhang Ming-Rong

2-((1E,3E)-4-(6-(11C-methylamino)pyridin-3-yl)buta-1,3-dienyl)benzo[d]thiazol-6-ol (11C-PBB3) is a clinically useful PET probe that we developed for in vivo imaging of tau pathology in the human brain. To ensure the availability of this probe among multiple PET facilities, in the present study we established protocols for the radiosynthesis and quality control of 11C-PBB3 and for the characterization of its photoisomerization, biodistribution, and metabolism. Methods: 11C-PBB3 was synthesized by reaction of the tert-butyldimethylsilyl desmethyl precursor (1) with 11C-methyl iodide using potassium hydroxide as a base, followed by deprotection. Photoisomerization of 11C-PBB3 under fluorescent light was determined. The biodistribution and metabolite analysis of 11C-PBB3 was determined in mice using the dissection method. Results: 11C-PBB3 was synthesized with 15.4% ± 2.8% radiochemical yield (decay-corrected, n = 50) based on the cyclotron-produced 11C-CO2 and showed an averaged synthesis time of 35 min from the end of bombardment. The radiochemical purity and specific activity of 11C-PBB3 were 98.0% ± 2.3% and 180.2 ± 44.3 GBq/μmol, respectively, at the end of synthesis (n = 50). 11C-PBB3 showed rapid photoisomerization, and its radiochemical purity decreased to approximately 50% at 10 min after exposure to fluorescent light. After the fluorescent light was switched off, 11C-PBB3 retained more than 95% radiochemical purity over 60 min. A suitable brain uptake (1.92% injected dose/g tissue) of radioactivity was observed at 1 min after the probe injection, which was followed by rapid washout from the brain tissue. More than 70% of total radioactivity in the mouse brain homogenate at 5 min after injection represented the unchanged 11C-PBB3, despite its rapid metabolism in the plasma. Conclusion: 11C-PBB3 was produced with sufficient radioactivity and high quality, demonstrating its clinical utility. The present results of radiosynthesis, photoisomerization, biodistribution, and metabolite analysis could be helpful for the reliable production and application of 11C-PBB3 in diverse PET facilities.


Applied Radiation and Isotopes | 2002

Development of an automated system for synthesizing 18F-labeled compounds using [18F]fluoroethyl bromide as a synthetic precursor

Ming-Rong Zhang; Akio Tsuchiyama; Terushi Haradahira; Yuichiro Yoshida; Kenji Furutsuka; Kazutoshi Suzuki

An automated system was developed to synthesize 18F-labeled compounds using [18F]fluoroethyl bromide ([18F]FEtBr) as a synthetic precursor. The apparatus makes possible the following sequence of processes: (1) production of an aqueous solution of [18F]fluoride ([18F]F-), (2) recovery of [18F]F- from target chamber, (3) drying of [18F]F-, (4) formation and distillation of [18F]FEtBr into a trapping vessel, (5) alkylation of target compounds with [18F]FEtBr, (6) High performance liquid chromatography purification and (7) formulation. [18F]FEtBr, the synthetic precursor for fluoroethylation, was labeled via nucleophilic displacement of 2-trifluoromethanesulfonyloxy ethylbromide (BrCH2CH2OTf) with [18F]F- and was purified from the reaction mixture by distillation. After the conditions for forming [18F]FEtBr and drying [18F]F- were optimized, [18F]FEtBr was obtained in a radiochemical yield of 71 +/- 13% (n = 21, based on [18F]F-, corrected for decay) and a radiochemical purity of 98 +/- 1.4% at end of the syntheses (EOS). Using this automated system, [18F]fluoroethylspiperone ([18F]FEtSP) was prepared by reacting spiperone with [18F]FEtBr in a radiochemical yield and purity of 56 +/- 12% (n = 5, based on [18F]FEtBr, corrected for decay) and 97 +/- 1.5% with a specific activity of 310 +/- 120 GBq/mumol at EOS. The total synthesis time was 55 +/- 2.3 min from the end of bombardment and the developed system has proved to be reliable and reproducible.


Nuclear Medicine and Biology | 2002

Synthesis and preliminary evaluation of [18F]FEtP4A, a promising PET tracer for mapping acetylcholinesterase in vivo

Ming-Rong Zhang; Akio Tsuchiyama; Terushi Haradahira; Kenji Furutsuka; Yuichiro Yoshida; Takayo Kida Junko Noguchi; Toshiaki Irie; Kazutoshi Suzuki

N-[18F]Fluoroethyl-4-piperidyl acetate ([18F]FEtP4A), an analog of [11C]MP4A for mapping brain acetylcholineseterase (AchE) activity, was prepared by reacting 4-piperidyl acetate (P4A) with [18F]fluoroethyl bromide ([18F]FEtBr) using a newly developed automated system. Preliminary evaluation showed that the initial uptake of [18F]FEtP4A in the mouse brain was > 8% injected dose/g tissue. The distribution pattern of [18F]FEtP4A in the brain was striatum>cerebral cortex>cerebellum within 10-120 min post-injection, which reflected the distribution rank pattern of AchE activity in the brain. Moreover, chemical analysis of in vivo radioactive metabolites in the mouse brain indicated that 83% of [18F]FEtP4A was hydrolyzed to N-[18F]fluoroethyl-4-piperidinol ([18F]FEtP4OH) after 1 min intravenous injection. From these results, [18F]FEtP4A may become a promising PET tracer for mapping the AchE in vivo.


Nuclear Medicine and Biology | 2015

Identification of a major radiometabolite of [11C]PBB3.

Hiroki Hashimoto; Kazunori Kawamura; Makoto Takei; Nobuyuki Igarashi; Tomoya Fujishiro; Satoshi Shiomi; Ryuji Watanabe; Masatoshi Muto; Kenji Furutsuka; Takehito Ito; Tomoteru Yamasaki; Joji Yui; Kazuyoshi Nemoto; Yasuyuki Kimura; Makoto Higuchi; Ming-Rong Zhang

INTRODUCTION [(11)C]PBB3 is a clinically used positron emission tomography (PET) probe for in vivo imaging of tau pathology in the brain. Our previous study showed that [(11)C]PBB3 was rapidly decomposed to a polar radiometabolite in the plasma of mice. For the pharmacokinetic evaluation of [(11)C]PBB3 it is important to elucidate the characteristics of radiometabolites. In this study, we identified the chemical structure of a major radiometabolite of [(11)C]PBB3 and proposed the metabolic pathway of [(11)C]PBB3. METHODS Carrier-added [(11)C]PBB3 was injected into a mouse for in vivo metabolite analysis. The chemical structure of a major radiometabolite was identified using LC-MS. Mouse and human liver microsomes and liver S9 samples were incubated with [(11)C]PBB3 in vitro. In silico prediction software was used to assist in the determination of the metabolite and metabolic pathway of [(11)C]PBB3. RESULTS In vivo analysis showed that the molecular weight of a major radiometabolite of [(11)C]PBB3, which was called as [(11)C]M2, was m/z 390 [M+H(+)]. In vitro analysis assisted by in silico prediction showed that [(11)C]M2, which was not generated by cytochrome P450 enzymes (CYPs), was generated by sulfated conjugation mediated by a sulfotransferase. CONCLUSION The major radiometabolite, [(11)C]M2, was identified as a sulfated conjugate of [(11)C]PBB3. [(11)C]PBB3 was metabolized mainly by a sulfotransferase and subsidiarily by CYPs.


Bioorganic & Medicinal Chemistry | 2011

Efficient synthesis and chiral separation of 11C-labeled ibuprofen assisted by DMSO for imaging of in vivo behavior of the individual isomers by positron emission tomography

Tatsuya Kikuchi; Maki Okada; Nobuki Nengaki; Kenji Furutsuka; Hidekatsu Wakizaka; Toshimitsu Okamura; Ming-Rong Zhang; Koichi Kato

The pharmacological mechanisms focusing on chiral isomer of ibuprofen are not fully understood. Only the (S)-isomer of ibuprofen inhibits cyclooxygenases, which mediates the generation of prostanoids and thromboxanes. Consequently, (S)-isomers represent a major promoter of the anti-inflammatory effect, and the effects of the (R)-isomers have not been widely discussed. However, more recently, the cyclooxygenase-independent pharmacological effects of ibuprofen have been elucidated. Pharmacokinetic studies with individual isomers of ibuprofen by positron emission tomography should aid our understanding of the pharmacological mechanisms of ibuprofen. The efficient (11)C-labeling of ibuprofen for chiral separation via the TBAF-promoted α-[(11)C]methylation was achieved by using DMSO rather than THF as the reaction solvent. The robust production of the radiochemically labile (11)C-labeled ibuprofen ester was realized by the protective effect of DMSO on radiolysis. After intravenous injection of each enantiomer of [(11)C]ibuprofen, significantly high radioactivity was observed in the joints of arthritis mice when compared to the levels observed in normal mice. However, the high accumulation was equivalent between the enantiomers, indicating that ibuprofen is accumulated in the arthritic joints regardless of the expression of cyclooxygenases.


Applied Radiation and Isotopes | 2011

High-yield automated synthesis of [18F]fluoroazomycin arabinoside ([18F]FAZA) for hypoxia-specific tumor imaging

Kazutaka Hayashi; Kenji Furutsuka; Makot Takei; Masatoshi Muto; Ryuji Nakao; Hatsumi Aki; Kazutoshi Suzuki; Toshimitsu Fukumura

The aim of this study was to develop an efficient fully automated synthesis method to achieve a high radiochemical yield of [(18)F]FAZA with a small amount of precursor. A small cartridge containing 25mg of the QMA resin was prepared and evaluated to obtain [(18)F]F(-) in a small quantity of base (K(2)CO(3)), which might allow the use of a small amount of precursor. The labeling and hydrolyzing conditions for [(18)F]FAZA synthesis were also investigated manually. No-carrier-added [(18)F]F(-) was trapped on the small QMA cartridge and eluted with a mixture of Krytofix 222 (2.26 mg, 6.0 μmol) and K(2)CO(3) (0.69 mg, 5.0 μmol) in 70% MeCN (0.4 mL). The automated synthesis of [(18)F]FAZA was optimally performed with a modified NIRS original synthesis system for clinical use, by labeling 2.5mg (5.2 μmol) of the precursor in DMSO (0.4 mL) at 120°C for 10 min, and then by hydrolyzing the (18)F-labeled intermediate with 0.1M NaOH (0.5 mL) at room temperature for 3 min. Using the above condition, the [(18)F]FAZA injection was obtained with a high radiochemical yield of 52.4±5.3% (decay-corrected, n=8) within 50.5±1.5 min.


Nuclear Medicine and Biology | 2014

Binding potential of (E)-[11C]ABP688 to metabotropic glutamate receptor subtype 5 is decreased by the inclusion of its 11C-labelled Z-isomer

Kazunori Kawamura; Tomoteru Yamasaki; Katsushi Kumata; Kenji Furutsuka; Makoto Takei; Hidekatsu Wakizaka; Masayuki Fujinaga; Kaori Kariya; Joji Yui; Akiko Hatori; Lin Xie; Yoko Shimoda; Hiroki Hashimoto; Kazutaka Hayashi; Ming-Rong Zhang

INTRODUCTION [(11)C]ABP688 is a promising positron emission tomography (PET) ligand for imaging of metabotropic glutamate receptor subtype 5 (mGlu5 receptor). Of the two geometric isomers of ABP688, (E)-ABP688 has a greater affinity towards mGlu5 receptors than (Z)-ABP688. Therefore, a high ratio of E-isomer is required when using [(11)C]ABP688 as a PET probe for imaging and quantification of mGlu5 receptors. The aim of this study was to evaluate the effect (Z)-[(11)C]ABP688 on the synthesis of [(11)C]ABP688 to be used for binding (E)-[(11)C]ABP688 in the brain. METHODS We synthesized and separated (E)- and (Z)-[(11)C]ABP688 by purification using an improved preparative high-performance liquid chromatography (HPLC) method equipped with a COSMOSIL Cholester column. We performed an in vitro binding assay in rat brain homogenates and PET studies of the rat brains using (E)- and (Z)-[(11)C]ABP688. RESULTS (E)- and (Z)-[(11)C]ABP688 were successfully obtained with suitable radioactivity for application. In the in vitro assay, the Kd value of (E)-[(11)C]ABP688 (5.7 nmol/L) was higher than that of (Z)-[(11)C]ABP688 (140 nmol/L). In the PET study of the rat brain, high radioactivity after injection of (E)-[(11)C]ABP688 was observed in regions rich in mGlu5 receptors such as the striatum and hippocampus. In contrast, after injection of (Z)-[(11)C]ABP688, radioactivity did not accumulate in the brain. Furthermore, BPND in the striatum and hippocampus was highly correlated (R(2) = 0.99) with the percentage of (E)-[(11)C]ABP688 of the total radioactivity of (E)- and (Z)-[(11)C]ABP688 in the injection. CONCLUSION We demonstrated that including (Z)-[(11)C]ABP688 in the [(11)C]ABP688 injection can decrease BPND in regions rich in mGlu5 receptors. Routine production of (E)-[(11)C]ABP688 will be helpful for imaging and quantification of mGlu5 receptors in clinical studies.


Bioorganic & Medicinal Chemistry | 2003

N-[18F]fluoroethyl-4-piperidyl acetate ([18F]FEtP4A): A PET tracer for imaging brain acetylcholinesterase in vivo.

Ming-Rong Zhang; Kenji Furutsuka; Jun Maeda; Tatsuya Kikuchi; Takayo Kida; Takashi Okauchi; Toshiaki Irie; Kazutoshi Suzuki

N-[(18)F]Fluoroethyl-4-piperidyl acetate ([(18)F]FEtP4A) was synthesized and evaluated as a PET tracer for imaging brain acetylcholinesterase (AchE) in vivo. [(18)F]FEtP4A was previously prepared by reacting 4-piperidyl acetate (P4A) with 2-[(18)F]fluoroethyl bromide ([(18)F]FEtBr) at 130 degrees C for 30 min in 37% radiochemical yield using an automated synthetic system. In this work, [(18)F]FEtP4A was synthesized by reacting P4A with 2-[(18)F]fluoroethyl iodide ([(18)F]FEtI) or 2-[(18)F]fluoroethyl triflate ([(18)F]FEtOTf in improved radiochemical yields, compared with [(18)F]FEtBr under the corresponding condition. Ex vivo autoradiogram of rat brain and PET summation image of monkey brain after iv injection of [(18)F]FEtP4A displayed a high radioactivity in the striatum, a region with the highest AchE activity in the brain. Moreover, the distribution pattern of (18)F radioactivity was consistent with that of AchE in the brain: striatum>frontal cortex>cerebellum. In the rat and monkey plasma, two radioactive metabolites were detected. However, their presence might not preclude the imaging studies for AchE in the brain, because they were too hydrophilic to pass the blood-brain barrier and to enter the brain. In the rat brain, only [(18)F]fluoroethyl-4-piperidinol ([(18)F]FEtP4OH) was detected at 30 min postinjection. The hydrolytic [(18)F]FEtP4OH displayed a slow washout and a long retention in the monkey brain until the PET experiment (120 min). Although [(18)F]FEtP4A is a potential PET tracer for imaging AchE in vivo, its lower hydrolytic rate and lower specificity for AchE than those of [(11)C]MP4A may limit its usefulness for the quantitative measurement for AchE in the primate brain.

Collaboration


Dive into the Kenji Furutsuka's collaboration.

Top Co-Authors

Avatar

Ming-Rong Zhang

National Institute of Radiological Sciences

View shared research outputs
Top Co-Authors

Avatar

Kazunori Kawamura

National Institute of Radiological Sciences

View shared research outputs
Top Co-Authors

Avatar

Kazutoshi Suzuki

National Institute of Radiological Sciences

View shared research outputs
Top Co-Authors

Avatar

Makoto Takei

National Institute of Radiological Sciences

View shared research outputs
Top Co-Authors

Avatar

Hiroki Hashimoto

National Institute of Radiological Sciences

View shared research outputs
Top Co-Authors

Avatar

Joji Yui

National Institute of Radiological Sciences

View shared research outputs
Top Co-Authors

Avatar

Tomoteru Yamasaki

National Institute of Radiological Sciences

View shared research outputs
Top Co-Authors

Avatar

Katsushi Kumata

National Institute of Radiological Sciences

View shared research outputs
Top Co-Authors

Avatar

Masatoshi Muto

National Institute of Radiological Sciences

View shared research outputs
Top Co-Authors

Avatar

Akiko Hatori

National Institute of Radiological Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge