Network


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

Hotspot


Dive into the research topics where Michiko Tawada is active.

Publication


Featured researches published by Michiko Tawada.


Journal of Medicinal Chemistry | 2013

Discovery of potent Mcl-1/Bcl-xL dual inhibitors by using a hybridization strategy based on structural analysis of target proteins.

Yuta Tanaka; Katsuji Aikawa; Goushi Nishida; Misaki Homma; Satoshi Sogabe; Shigeru Igaki; Yumi Hayano; Tomoya Sameshima; Ikuo Miyahisa; Tomohiro Kawamoto; Michiko Tawada; Yumi N. Imai; Masakazu Inazuka; Nobuo Cho; Yasuhiro Imaeda; Tomoyasu Ishikawa

Mcl-1 and Bcl-xL are crucial regulators of apoptosis, therefore dual inhibitors of both proteins could serve as promising new anticancer drugs. To design Mcl-1/Bcl-xL dual inhibitors, we performed structure-guided analyses of the corresponding selective Mcl-1 and Bcl-xL inhibitors. A cocrystal structure of a pyrazolo[1,5-a]pyridine derivative with Mcl-1 protein was successfully determined and revealed the protein-ligand binding mode. The key structure for Bcl-xL inhibition was further confirmed through the substructural analysis of ABT-263, a representative Bcl-xL/Bcl-2/Bcl-w inhibitor developed by Abbott Laboratories. On the basis of the structural data from this analysis, we designed hybrid compounds by tethering the Mcl-1 and Bcl-xL inhibitors together. The results of X-ray crystallographic analysis of hybrid compound 10 in complexes with both Mcl-1 and Bcl-xL demonstrated its binding mode with each protein. Following further optimization, compound 11 showed potent Mcl-1/Bcl-xL dual inhibitory activity (Mcl-1, IC50 = 0.088 μM; and Bcl-xL, IC50 = 0.0037 μM).


Bioorganic & Medicinal Chemistry | 2011

Identification of 3-aminomethyl-1,2-dihydro-4-phenyl-1-isoquinolones: a new class of potent, selective, and orally active non-peptide dipeptidyl peptidase IV inhibitors that form a unique interaction with Lys554.

Yoshihiro Banno; Yasufumi Miyamoto; Mitsuru Sasaki; Satoru Oi; Tomoko Asakawa; Osamu Kataoka; Koji Takeuchi; Nobuhiro Suzuki; Koji Ikedo; Takuo Kosaka; Shigetoshi Tsubotani; Akiyoshi Tani; Miyuki Funami; Michiko Tawada; Yoshio Yamamoto; Kathleen Aertgeerts; Jason Yano; Hironobu Maezaki

The design, synthesis, and structure-activity relationships of a new class of potent and orally active non-peptide dipeptidyl peptidase IV (DPP-4) inhibitors, 3-aminomethyl-1,2-dihydro-4-phenyl-1-isoquinolones, are described. We hypothesized that the 4-phenyl group of the isoquinolone occupies the S1 pocket of the enzyme, the 3-aminomethyl group forms an electrostatic interaction with the S2 pocket, and the introduction of a hydrogen bond donor onto the 6- or 7-substituent provides interaction with the hydrophilic region of the enzyme. Based on this hypothesis, intensive research focused on developing new non-peptide DPP-4 inhibitors has been carried out. Among the compounds designed in this study, we identified 2-[(3-aminomethyl-2-(2-methylpropyl)-1-oxo-4-phenyl-1,2-dihydro-6-isoquinolinyl)oxy]acetamide (35a) as a potent, selective, and orally bioavailable DPP-4 inhibitor, which exhibited in vivo efficacy in diabetic model rats. Finally, X-ray crystallography of 35a in a complex with the enzyme validated our hypothesized binding mode and identified Lys554 as a new target-binding site available for DPP-4 inhibitors.


Bioorganic & Medicinal Chemistry | 2013

Design, synthesis, and evaluation of novel VEGFR2 kinase inhibitors: Discovery of [1,2,4]triazolo[1,5-a]pyridine derivatives with slow dissociation kinetics

Yuya Oguro; Douglas R. Cary; Naoki Miyamoto; Michiko Tawada; Hidehisa Iwata; Hiroshi Miki; Akira Hori; Shinichi Imamura

For the purpose of discovering novel type-II inhibitors of vascular endothelial growth factor receptor 2 (VEGFR2) kinase, we designed and synthesized 5,6-fused heterocyclic compounds bearing a anilide group. A co-crystal structure analysis of imidazo[1,2-b]pyridazine derivative 2 with VEGFR2 revealed that the N1-nitrogen of imidazo[1,2-b]pyridazine core interacts with the backbone NH group of Cys919. To retain this essential interaction, we designed a series of imidazo[1,2-a]pyridine, [1,2,4]triazolo[1,5-a]pyridine, thiazolo[5,4-b]pyridine, and 1,3-benzothiazole derivatives maintaining a ring nitrogen as hydrogen bond acceptor (HBA) at the corresponding position. All compounds thus designed displayed strong inhibitory activity against VEGFR2 kinase, and the [1,2,4]triazolo[1,5-a]pyridine 13d displayed favorable physicochemical properties. Furthermore, 13d inhibited VEGFR2 kinase with slow dissociation kinetics and also inhibited platelet-derived growth factor receptor (PDGFR) kinases. Oral administration of 13d showed potent anti-tumor efficacy in DU145 and A549 xenograft models in nude mice.


Bioorganic & Medicinal Chemistry | 2013

Structure-based design, synthesis, and evaluation of imidazo[1,2-b]pyridazine and imidazo[1,2-a]pyridine derivatives as novel dual c-Met and VEGFR2 kinase inhibitors.

Shigemitsu Matsumoto; Naoki Miyamoto; Takaharu Hirayama; Hideyuki Oki; Kengo Okada; Michiko Tawada; Hidehisa Iwata; Kazuhide Nakamura; Seiji Yamasaki; Hiroshi Miki; Akira Hori; Shinichi Imamura

To identify compounds with potent antitumor efficacy for various human cancers, we aimed to synthesize compounds that could inhibit c-mesenchymal epithelial transition factor (c-Met) and vascular endothelial growth factor receptor 2 (VEGFR2) kinases. We designed para-substituted inhibitors by using co-crystal structural information from c-Met and VEGFR2 in complex with known inhibitors. This led to the identification of compounds 3a and 3b, which were capable of suppressing both c-Met and VEGFR2 kinase activities. Further optimization resulted in pyrazolone and pyridone derivatives, which could form intramolecular hydrogen bonds to enforce a rigid conformation, thereby producing potent inhibition. One compound of particular note was the imidazo[1,2-a]pyridine derivative (26) bearing a 6-methylpyridone ring, which strongly inhibited both c-Met and VEGFR2 enzyme activities (IC50=1.9, 2.2 nM), as well as proliferation of c-Met-addicted MKN45 cells and VEGF-stimulated human umbilical vein endothelial cells (IC50=5.0, 1.8 nM). Compound 26 exhibited dose-dependent antitumor efficacy in vivo in MKN45 (treated/control ratio [T/C]=4%, po, 5mg/kg, once-daily) and COLO205 (T/C=13%, po, 15 mg/kg, once-daily) mouse xenograft models.


Bioorganic & Medicinal Chemistry | 2011

Discovery of potent, selective, and orally bioavailable quinoline-based dipeptidyl peptidase IV inhibitors targeting Lys554.

Hironobu Maezaki; Yoshihiro Banno; Yasufumi Miyamoto; Yuusuke Moritou; Tomoko Asakawa; Osamu Kataoka; Koji Takeuchi; Nobuhiro Suzuki; Koji Ikedo; Takuo Kosaka; Masako Sasaki; Shigetoshi Tsubotani; Akiyoshi Tani; Miyuki Funami; Yoshio Yamamoto; Michiko Tawada; Kathleen Aertgeerts; Jason Yano; Satoru Oi

Dipeptidyl peptidase IV (DPP-4) inhibition is a validated therapeutic option for type 2 diabetes, exhibiting multiple antidiabetic effects with little or no risk of hypoglycemia. In our studies involving non-covalent DPP-4 inhibitors, a novel series of quinoline-based inhibitors were designed based on the co-crystal structure of isoquinolone 2 in complex with DPP-4 to target the side chain of Lys554. Synthesis and evaluation of designed compounds revealed 1-[3-(aminomethyl)-4-(4-methylphenyl)-2-(2-methylpropyl)quinolin-6-yl]piperazine-2,5-dione (1) as a potent, selective, and orally active DPP-4 inhibitor (IC₅₀=1.3 nM) with long-lasting ex vivo activity in dogs and excellent antihyperglycemic effects in rats. A docking study of compound 1 revealed a hydrogen-bonding interaction with the side chain of Lys554, suggesting this residue as a potential target site useful for enhancing DPP-4 inhibition.


Bioorganic & Medicinal Chemistry Letters | 2012

Symmetrical approach of spiro-pyrazolidinediones as acetyl-CoA carboxylase inhibitors.

Makoto Kamata; Tohru Yamashita; Asato Kina; Michiko Tawada; Satoshi Endo; Atsushi Mizukami; Masako Sasaki; Akiyoshi Tani; Yoshihide Nakano; Yuuki Watanabe; Naoki Furuyama; Miyuki Funami; Nobuyuki Amano; Kohji Fukatsu

Spiro-pyrazolidinedione derivatives without quaternary chiral center were discovered by structure-based drug design and characterized as potent acetyl-CoA carboxylase (ACC) inhibitors. The high metabolic stability of the spiro-pyrazolo[1,2-a]pyridazine scaffold and enhancement of the activity by incorporation of a 7-methoxy group on the benzothiophene core successfully led to the identification of compound 4c as an orally bioavailable and highly potent ACC inhibitor. Oral administration of 4c significantly decreased the values of the respiratory quotient in rats, indicating the stimulation of fatty acid oxidation.


Bioorganic & Medicinal Chemistry | 2011

Melanin-concentrating hormone receptor 1 antagonists: synthesis, structure-activity relationship, docking studies, and biological evaluation of 2,3,4,5-tetrahydro-1H-3-benzazepine derivatives.

Shizuo Kasai; Masahiro Kamaura; Makoto Kamata; Kazuyoshi Aso; Hitomi Ogino; Yoshihide Nakano; Kaoru Watanabe; Tomoko Kaisho; Michiko Tawada; Yasutaka Nagisa; Shiro Takekawa; Koki Kato; Nobuhiro Suzuki; Yuji Ishihara

Melanin-concentrating hormone receptor 1 (MCHR1) antagonists have been studied as potential agents for the treatment of obesity. Initial structure-activity relationship studies of in-house hit compound 1a and subsequent optimization studies resulted in the identification of tetrahydroisoquinoline derivative 23, 1-(2-acetyl-1,2,3,4-tetrahydroisoquinolin-7-yl)-4-[4-(4-chlorophenyl)piperidin-1-yl]butan-1-one, as a potent hMCHR1 antagonist. A homology model of hMCHR1 suggests that these compounds interact with Asn 294 and Asp 123 in the binding site of hMCHR1 to enhance binding affinity. Oral administration of compound 23 dose-dependently reduced food intake in diet-induced obesity (DIO)-F344 rats.


Oncotarget | 2018

TP-064, a potent and selective small molecule inhibitor of PRMT4 for multiple myeloma

Kazuhide Nakayama; Magdalena M. Szewczyk; Carlo dela Sena; Hong Wu; Aiping Dong; Hong Zeng; Fengling Li; Renato Ferreira de Freitas; Mohammad S. Eram; Matthieu Schapira; Yuji Baba; Mihoko Kunitomo; Douglas R. Cary; Michiko Tawada; Akihiro Ohashi; Yasuhiro Imaeda; Kumar Singh Saikatendu; Charles E. Grimshaw; Masoud Vedadi; Cheryl H. Arrowsmith; Dalia Barsyte-Lovejoy; Atsushi Kiba; Daisuke Tomita; Peter J. Brown

Protein arginine methyltransferase (PRMT) 4 (also known as coactivator-associated arginine methyltransferase 1; CARM1) is involved in a variety of biological processes and is considered as a candidate oncogene owing to its overexpression in several types of cancer. Selective PRMT4 inhibitors are useful tools for clarifying the molecular events regulated by PRMT4 and for validating PRMT4 as a therapeutic target. Here, we report the discovery of TP-064, a potent, selective, and cell-active chemical probe of human PRMT4 and its co-crystal structure with PRMT4. TP-064 inhibited the methyltransferase activity of PRMT4 with high potency (half-maximal inhibitory concentration, IC50 < 10 nM) and selectivity over other PRMT family proteins, and reduced arginine dimethylation of the PRMT4 substrates BRG1-associated factor 155 (BAF155; IC50= 340 ± 30 nM) and Mediator complex subunit 12 (MED12; IC50 = 43 ± 10 nM). TP-064 treatment inhibited the proliferation of a subset of multiple myeloma cell lines, with affected cells arrested in G1 phase of the cell cycle. TP-064 and its negative control (TP-064N) will be valuable tools to further investigate the biology of PRMT4 and the therapeutic potential of PRMT4 inhibition.


Bioorganic & Medicinal Chemistry | 2018

Discovery of orally efficacious RORγt inverse agonists. Part 2: Design, synthesis, and biological evaluation of novel tetrahydroisoquinoline derivatives

Mitsunori Kono; Tsuneo Oda; Michiko Tawada; Takashi Imada; Yoshihiro Banno; Naohiro Taya; Tetsuji Kawamoto; Hidekazu Tokuhara; Yoshihide Tomata; Naoki Ishii; Atsuko Ochida; Yoshiyuki Fukase; Tomoya Yukawa; Shoji Fukumoto; Hiroyuki Watanabe; Keiko Uga; Akira Shibata; Hideyuki Nakagawa; Mikio Shirasaki; Yasushi Fujitani; Masashi Yamasaki; Junya Shirai; Satoshi Yamamoto

A series of tetrahydroisoquinoline derivatives were designed, synthesized, and evaluated for their potential as novel orally efficacious retinoic acid receptor-related orphan receptor-gamma t (RORγt) inverse agonists for the treatment of Th17-driven autoimmune diseases. We carried out cyclization of the phenylglycinamide core by structure-based drug design and successfully identified a tetrahydroisoquinoline carboxylic acid derivative 14 with good biochemical binding and cellular reporter activity. Interestingly, the combination of a carboxylic acid tether and a central fused bicyclic ring was crucial for optimizing PK properties, and the compound 14 showed significantly improved PK profile. Successive optimization of the carboxylate tether led to the discovery of compound 15 with increased inverse agonistic activity and an excellent PK profile. Oral treatment of mice with compound 15 robustly and dose-dependently inhibited IL-17A production in an IL23-induced gene expression assay.


Bioorganic & Medicinal Chemistry | 2016

Novel approach of fragment-based lead discovery applied to renin inhibitors

Michiko Tawada; Shinkichi Suzuki; Yasuhiro Imaeda; Hideyuki Oki; Gyorgy Snell; Craig A. Behnke; Mitsuyo Kondo; Naoki Tarui; Toshimasa Tanaka; Takanobu Kuroita; Masaki Tomimoto

A novel approach was conducted for fragment-based lead discovery and applied to renin inhibitors. The biochemical screening of a fragment library against renin provided the hit fragment which showed a characteristic interaction pattern with the target protein. The hit fragment bound only to the S1, S3, and S3SP (S3 subpocket) sites without any interactions with the catalytic aspartate residues (Asp32 and Asp215 (pepsin numbering)). Prior to making chemical modifications to the hit fragment, we first identified its essential binding sites by utilizing the hit fragments substructures. Second, we created a new and smaller scaffold, which better occupied the identified essential S3 and S3SP sites, by utilizing library synthesis with high-throughput chemistry. We then revisited the S1 site and efficiently explored a good building block attaching to the scaffold with library synthesis. In the library syntheses, the binding modes of each pivotal compound were determined and confirmed by X-ray crystallography and the library was strategically designed by structure-based computational approach not only to obtain a more active compound but also to obtain informative Structure Activity Relationship (SAR). As a result, we obtained a lead compound offering synthetic accessibility as well as the improved in vitro ADMET profiles. The fragments and compounds possessing a characteristic interaction pattern provided new structural insights into renins active site and the potential to create a new generation of renin inhibitors. In addition, we demonstrated our FBDD strategy integrating highly sensitive biochemical assay, X-ray crystallography, and high-throughput synthesis and in silico library design aimed at fragment morphing at the initial stage was effective to elucidate a pocket profile and a promising lead compound.

Collaboration


Dive into the Michiko Tawada's collaboration.

Top Co-Authors

Avatar

Akiyoshi Tani

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hironobu Maezaki

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Masako Sasaki

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Miyuki Funami

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Yasuhiro Imaeda

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Yoshihiro Banno

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Koji Ikedo

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Makoto Kamata

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Satoru Oi

Takeda Pharmaceutical Company

View shared research outputs
Researchain Logo
Decentralizing Knowledge