Network


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

Hotspot


Dive into the research topics where Shinichi Masada is active.

Publication


Featured researches published by Shinichi Masada.


Journal of Medicinal Chemistry | 2012

Melanin-concentrating hormone receptor 1 antagonists. Synthesis and structure-activity relationships of novel 3-(aminomethyl)quinolines.

Makoto Kamata; Toshiro Yamashita; Toshihiro Imaeda; Toshio Tanaka; Shinichi Masada; Masahiro Kamaura; Shizuo Kasai; Ryoma Hara; Shigekazu Sasaki; Shiro Takekawa; Asano Asami; Tomoko Kaisho; Nobuhiro Suzuki; Shuntaro Ashina; Hitomi Ogino; Yoshihide Nakano; Yasutaka Nagisa; Koki Kato; Kaneyoshi Kato; Yuji Ishihara

It was found that 3-(aminomethyl)quinoline derivatives showed high binding affinities for melanin-concentrating hormone receptor 1 (MCHR1) with reduced affinity for serotonin receptor 2c (5-HT2c) when the dihydronaphthalene nucleus of compound 1 (human MCHR1, IC(50) = 1.9 nM; human 5-HT2c receptor, IC(50) = 0.53 nM) was replaced by other bicyclic core scaffolds. Among the synthesized compounds, 8-methylquinoline derivative 5v especially showed high binding affinity (IC(50) = 0.54 nM), potent in vitro antagonistic activity (IC(50) = 2.8 nM) for MCHR1, and negligible affinity for 5-HT2c receptor (IC(50) > 1000 nM). Oral administration of 5v significantly and dose-dependently suppressed nocturnal food intake in diet-induced obese rats and did not affect food intake in MCHR1-deficient mice. These results and rat pharmacokinetic study findings suggested that compound 5v is a highly potent, orally bioavailable, and centrally acting nonpeptide MCHR1 antagonist.


Journal of Medicinal Chemistry | 2012

Synthesis, Structure–Activity Relationship, and Pharmacological Studies of Novel Melanin-Concentrating Hormone Receptor 1 Antagonists 3-Aminomethylquinolines: Reducing Human Ether-a-go-go-Related Gene (hERG) Associated Liabilities

Shizuo Kasai; Makoto Kamata; Shinichi Masada; Jun Kunitomo; Masahiro Kamaura; Tomohiro Okawa; Kazuaki Takami; Hitomi Ogino; Yoshihide Nakano; Shuntarou Ashina; Kaoru Watanabe; Tomoko Kaisho; Yumi N. Imai; Sunghi Ryu; Masaharu Nakayama; Yasutaka Nagisa; Shiro Takekawa; Koki Kato; Toshiki Murata; Nobuhiro Suzuki; Yuji Ishihara

Recently, we discovered 3-aminomethylquinoline derivative 1, a selective, highly potent, centrally acting, and orally bioavailable human MCH receptor 1 (hMCHR1) antagonist, that inhibited food intake in F344 rats with diet-induced obesity (DIO). Subsequent investigation of 1 was discontinued because 1 showed potent hERG K(+) channel inhibition in a patch-clamp study. To decrease hERG K(+) channel inhibition, experiments with ligand-based drug designs based on 1 and a docking study were conducted. Replacement of the terminal p-fluorophenyl group with a cyclopropylmethoxy group, methyl group introduction on the benzylic carbon at the 3-position of the quinoline core, and employment of a [2-(acetylamino)ethyl]amino group as the amine portion eliminated hERG K(+) channel inhibitory activity in a patch-clamp study, leading to the discovery of N-{3-[(1R)-1-{[2-(acetylamino)ethyl]amino}ethyl]-8-methylquinolin-7-yl}-4-(cyclopropylmethoxy)benzamide (R)-10h. The compound (R)-10h showed potent inhibitory activity against hMCHR1 and dose-dependently suppressed food intake in a 2-day study on DIO-F344 rats. Furthermore, practical chiral synthesis of (R)-10h was performed to determine the molecules absolute configuration.


Bioorganic & Medicinal Chemistry | 2018

Discovery of orally efficacious RORγt inverse agonists, part 1: Identification of novel phenylglycinamides as lead scaffolds

Junya Shirai; Yoshihide Tomata; Mitsunori Kono; Atsuko Ochida; Yoshiyuki Fukase; Ayumu Sato; Shinichi Masada; Tetsuji Kawamoto; Kazuko Yonemori; Ryoukichi Koyama; Hideyuki Nakagawa; Masaharu Nakayama; Keiko Uga; Akira Shibata; Keiko Koga; Toshitake Okui; Mikio Shirasaki; Robert J. Skene; Bi-Ching Sang; Isaac D. Hoffman; Wes Lane; Yasushi Fujitani; Masashi Yamasaki; Satoshi Yamamoto

A series of novel phenylglycinamides as retinoic acid receptor-related orphan receptor-gamma t (RORγt) inverse agonists were discovered through optimization of a high-throughput screen hit 1. (R)-N-(2-((3,5-Difluoro-4-(trimethylsilyl)phenyl) amino)-1-(4-methoxyphenyl)-2-oxoethyl)-3-hydroxy-N-methylisoxazole-5-carboxamide (22) was identified as one of the best of these compounds. It displayed higher subtype selectivity and specificity over other nuclear receptors and demonstrated in vivo potency to suppress the transcriptional activity of RORγt in a mouse PD (pharmacodynamic) model upon oral administration.


Journal of Medicinal Chemistry | 2017

Discovery of 3,5-Diphenyl-4-methyl-1,3-oxazolidin-2-ones as Novel, Potent, and Orally Available Δ-5 Desaturase (D5D) Inhibitors

Jun Fujimoto; Rei Okamoto; Naoyoshi Noguchi; Ryoma Hara; Shinichi Masada; Tetsuji Kawamoto; Hiroki Nagase; Yumiko Okano Tamura; Mitsuaki Imanishi; Shuichi Takagahara; Kazuki Kubo; Kimio Tohyama; Koichi Iida; Tomohiro Andou; Ikuo Miyahisa; Junji Matsui; Ryouta Hayashi; Tsuyoshi Maekawa; Nobuyuki Matsunaga

The discovery and optimization of Δ-5 desaturase (D5D) inhibitors are described. Investigation of the 1,3-oxazolidin-2-one scaffold was inspired by a pharmacophore model constructed from the common features of several hit compounds, resulting in the identification of 3,5-diphenyl-1,3-oxazolidin-2-one 5h as a novel lead showing potent in vitro activity. Subsequent optimization focused on the modification of two metabolic sites, which provided (4S,5S)-5i, a derivative with improved metabolic stability. Moreover, adding a substituent into the upper phenyl moiety further enhanced the intrinsic activity, which led to the discovery of 5-[(4S,5S)-5-(4fluorophenyl)-4-methyl-2-oxo-1,3-oxazolidin-3-yl]benzene-1,3-dicarbonitrile (4S,5S)-5n, endowed with excellent D5D binding affinity, cellular activity, and high oral bioavailability in a mouse. It exhibited robust in vivo hepatic arachidonic acid/dihomo-γ-linolenic acid ratio reduction (a target engagement marker) in an atherosclerosis mouse model. Finally, an asymmetric synthetic procedure for this compound was established.


Archive | 1999

Oxadiazoline derivatives and their use as insecticides

Yasuyuki Kando; Makoto Noguchi; Atsuo Akayama; Shinichi Masada; Toshiyuki Kiji


Archive | 1999

Oxadiazoline derivative and their use as insecticides

Yasuyuki Kando; Makota Noguchi; Akayama Atsuo; Shinichi Masada; Toshiyuki Kiji


Journal of Medicinal Chemistry | 2018

Discovery of [cis-3-({(5R)-5-[(7-Fluoro-1,1-dimethyl-2,3-dihydro-1H-inden-5-yl)carbamoyl]-2-methoxy-7,8-dihydro-1,6-naphthyridin-6(5H)-yl}carbonyl)cyclobutyl]acetic Acid (TAK-828F) as a Potent, Selective, and Orally Available Novel Retinoic Acid Receptor-Related Orphan Receptor γt Inverse Agonist

Mitsunori Kono; Atsuko Ochida; Tsuneo Oda; Takashi Imada; Yoshihiro Banno; Naohiro Taya; Shinichi Masada; Tetsuji Kawamoto; Kazuko Yonemori; Yoshi Nara; Yoshiyuki Fukase; Tomoya Yukawa; Hidekazu Tokuhara; Robert J. Skene; Bi-Ching Sang; Isaac D. Hoffman; Gyorgy Snell; Keiko Uga; Akira Shibata; Keiko Igaki; Yoshiki Nakamura; Hideyuki Nakagawa; Noboru Tsuchimori; Masashi Yamasaki; Junya Shirai; Satoshi Yamamoto


Tetrahedron | 2017

Direct and practical synthesis of 2′-O,4′-C-aminomethylene-bridged nucleic acid purine derivatives by transglycosylation

Tadashi Umemoto; Shinichi Masada; Kenichi Miyata; Mari Ogasawara-Shimizu; Shumpei Murata; Kazunori Nishi; Kazuhiro Ogi; Yoji Hayase; Nobuo Cho


Archive | 2013

Acide nucléique modifié inédit

Shumpei Murata; 俊平 村田; Shinichi Masada; 眞一 政田; N. Miyamoto; 宮本 直樹; Tadashi Umemoto; 忠士 梅本


Archive | 1999

Oxadiazolinderivate und ihre verwendung als insektizide Oxadiazolinderivate and their use as insecticidal

Yasuyuki Kando; Makoto Noguchi; Atsuo Akayama; Shinichi Masada; Toshiyuki Kiji

Collaboration


Dive into the Shinichi Masada's collaboration.

Top Co-Authors

Avatar

Toshiyuki Kiji

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Yasuyuki Kando

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Atsuo Akayama

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Makoto Noguchi

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Tetsuji Kawamoto

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Akira Shibata

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Atsuko Ochida

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Bi-Ching Sang

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Hideyuki Nakagawa

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Hitomi Ogino

Takeda Pharmaceutical Company

View shared research outputs
Researchain Logo
Decentralizing Knowledge