Network


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

Hotspot


Dive into the research topics where Bi-Ching Sang is active.

Publication


Featured researches published by Bi-Ching Sang.


Structure | 2002

Structures of the Cancer Related Aurora-A, FAK and EphA2 Protein Kinases from Nanovolume Crystallography

Jacek Nowakowski; Ciarán N. Cronin; Duncan E. McRee; Mark W. Knuth; Christian G. Nelson; Nikola P. Pavletich; Joe Rogers; Bi-Ching Sang; Daniel Scheibe; Ronald V. Swanson; Devon A. Thompson

Protein kinases are important drug targets in human cancers, inflammation, and metabolic diseases. This report presents the structures of kinase domains for three cancer-associated protein kinases: ephrin receptor A2 (EphA2), focal adhesion kinase (FAK), and Aurora-A. The expression profiles of EphA2, FAK, and Aurora-A in carcinomas suggest that inhibitors of these kinases may have inherent potential as therapeutic agents. The structures were determined from crystals grown in nanovolume droplets, which produced high-resolution diffraction data at 1.7, 1.9, and 2.3 A for FAK, Aurora-A, and EphA2, respectively. The FAK and Aurora-A structures are the first determined within two unique subfamilies of human kinases, and all three structures provide new insights into kinase regulation and the design of selective inhibitors.


Protein Science | 2004

Crystal structure of human dipeptidyl peptidase IV in complex with a decapeptide reveals details on substrate specificity and tetrahedral intermediate formation

Kathleen Aertgeerts; Sheng Ye; Mike Tennant; Michelle L. Kraus; Joe Rogers; Bi-Ching Sang; Robert J. Skene; David R. Webb; G. Sridhar Prasad

Dipeptidyl peptidase IV (DPPIV) is a member of the prolyl oligopeptidase family of serine proteases. DPPIV removes dipeptides from the N terminus of substrates, including many chemokines, neuropeptides, and peptide hormones. Specific inhibition of DPPIV is being investigated in human trials for the treatment of type II diabetes. To understand better the molecular determinants that underlie enzyme catalysis and substrate specificity, we report the crystal structures of DPPIV in the free form and in complex with the first 10 residues of the physiological substrate, Neuropeptide Y (residues 1–10; tNPY). The crystal structure of the free form of the enzyme reveals two potential channels through which substrates could access the active site—a so‐called propeller opening, and side opening. The crystal structure of the DPPIV/tNPY complex suggests that bioactive peptides utilize the side opening unique to DPPIV to access the active site. Other structural features in the active site such as the presence of a Glu motif, a well‐defined hydrophobic S1 subsite, and minimal long‐range interactions explain the substrate recognition and binding properties of DPPIV. Moreover, in the DPPIV/tNPY complex structure, the peptide is not cleaved but trapped in a tetrahedral intermediate that occurs during catalysis. Conformational changes of S630 and H740 between DPPIV in its free form and in complex with tNPY were observed and contribute to the stabilization of the tetrahedral intermediate. Our results facilitate the design of potent, selective small molecule inhibitors of DPPIV that may yield compounds for the development of novel drugs to treat type II diabetes.


Molecular Cancer Therapeutics | 2013

Biological Characterization of TAK-901, an Investigational, Novel, Multitargeted Aurora B Kinase Inhibitor

Pamela Farrell; Lihong Shi; Jennifer Matuszkiewicz; Deepika Balakrishna; Takashi Hoshino; Lilly Zhang; Sarah Elliott; Robyn Fabrey; Bumsup Lee; Petro Halkowycz; Bi-Ching Sang; Seigo Ishino; Toshiyuki Nomura; Mika Teratani; Yoshikazu Ohta; Charles E. Grimshaw; Bheema R. Paraselli; Takashi Satou; Ron de Jong

Protein kinases Aurora A, B, and C play essential roles during mitosis and cell division, are frequently elevated in cancer, and represent attractive targets for therapeutic intervention. TAK-901 is an investigational, multitargeted Aurora B kinase inhibitor derived from a novel azacarboline kinase hinge-binder chemotype. TAK-901 exhibited time-dependent, tight-binding inhibition of Aurora B, but not Aurora A. Consistent with Aurora B inhibition, TAK-901 suppressed cellular histone H3 phosphorylation and induced polyploidy. In various human cancer cell lines, TAK-901 inhibited cell proliferation with effective concentration values from 40 to 500 nmol/L. Examination of a broad panel of kinases in biochemical assays revealed inhibition of multiple kinases. However, TAK-901 potently inhibited only a few kinases other than Aurora B in intact cells, including FLT3 and FGFR2. In rodent xenografts, TAK-901 exhibited potent activity against multiple human solid tumor types, and complete regression was observed in the ovarian cancer A2780 model. TAK-901 also displayed potent activity against several leukemia models. In vivo biomarker studies showed that TAK-901 induced pharmacodynamic responses consistent with Aurora B inhibition and correlating with retention of TAK-901 in tumor tissue. These preclinical data highlight the therapeutic potential of TAK-901, which has entered phase I clinical trials in patients within a diverse range of cancers. Mol Cancer Ther; 12(4); 460–70. ©2013 AACR.


Bioorganic & Medicinal Chemistry | 2016

Discovery and optimization of 1,7-disubstituted-2,2-dimethyl-2,3-dihydroquinazolin-4(1 H )-ones as potent and selective PKC θ inhibitors

Taisuke Katoh; Takafumi Takai; Takafumi Yukawa; Tetsuya Tsukamoto; Etsurou Watanabe; Hideyuki Mototani; Takeo Arita; Hiroki Hayashi; Hideyuki Nakagawa; Michael G. Klein; Hua Zou; Bi-Ching Sang; Gyorgy Snell; Yoshihisa Nakada

A high-throughput screening campaign helped us to identify an initial lead compound (1) as a protein kinase C-θ (PKCθ) inhibitor. Using the docking model of compound 1 bound to PKCθ as a model, structure-based drug design was employed and two regions were identified that could be explored for further optimization, i.e., (a) a hydrophilic region around Thr442, unique to PKC family, in the inner part of the hinge region, and (b) a lipophilic region at the forefront of the ethyl moiety. Optimization of the hinge binder led us to find 1,3-dihydro-2H-imidazo[4,5-b]pyridin-2-one as a potent and selective hinge binder, which resulted in the discovery of compound 5. Filling the lipophilic region with a suitable lipophilic substituent boosted PKCθ inhibitory activity and led to the identification of compound 10. The co-crystal structure of compound 10 bound to PKCθ confirmed that both the hydrophilic and lipophilic regions were fully utilized. Further optimization of compound 10 led us to compound 14, which demonstrated an improved pharmacokinetic profile and inhibition of IL-2 production in a mouse.


Bioorganic & Medicinal Chemistry | 2017

Identification of novel quinazolinedione derivatives as RORγt inverse agonist

Yoshiyuki Fukase; Ayumu Sato; Yoshihide Tomata; Atsuko Ochida; Mitsunori Kono; Kazuko Yonemori; Keiko Koga; Toshitake Okui; Masashi Yamasaki; Yasushi Fujitani; Hideyuki Nakagawa; Ryoukichi Koyama; Masaharu Nakayama; Robert J. Skene; Bi-Ching Sang; Isaac D. Hoffman; Junya Shirai; Satoshi Yamamoto

Novel small molecules were synthesized and evaluated as retinoic acid receptor-related orphan receptor-gamma t (RORγt) inverse agonists for the treatment of inflammatory and autoimmune diseases. A hit compound, 1, was discovered by high-throughput screening of our compound library. The structure-activity relationship (SAR) study of compound 1 showed that the introduction of a chlorine group at the 3-position of 4-cyanophenyl moiety increased the potency and a 3-methylpentane-1,5-diamide linker is favorable for the activity. The carbazole moiety of 1 was also optimized; a quinazolinedione derivative 18i suppressed the increase of IL-17A mRNA level in the lymph node of a rat model of experimental autoimmune encephalomyelitis (EAE) upon oral administration. These results indicate that the novel quinazolinedione derivatives have great potential as orally available small-molecule RORγt inverse agonists for the treatment of Th17-driven autoimmune diseases. A U-shaped bioactive conformation of this chemotype with RORγt protein was also observed.


Pharmacology | 2018

Biochemical Properties of TAK-828F, a Potent and Selective Retinoid-Related Orphan Receptor Gamma t Inverse Agonist

Hideyuki Nakagawa; Ryoukichi Koyama; Yusuke Kamada; Atsuko Ochida; Mitsunori Kono; Junya Shirai; Satoshi Yamamoto; Geza Ambrus-Aikelin; Bi-Ching Sang; Masaharu Nakayama

Background/Aims: Retinoid-related orphan receptor gamma t (RORγt) is a master regulator of T helper 17 cells that plays a pivotal role in the production of inflammatory cytokines including interleukin (IL)-17. Therefore, RORγt has attracted much attention as a target receptor for the treatment of inflammatory diseases including rheumatoid arthritis, multiple sclerosis, inflammatory bowel diseases, and psoriasis. This study aims to characterize TAK-828F, a potent and selective RORγt inverse agonist. Methods: The biochemical properties of TAK-828F were evaluated using Time-Resolved Fluorescence Resonance Energy Transfer (TR-FRET) binding assay, surface plasmon resonance (SPR) biosensor assay, cofactor recruitment assay, reporter assay, and IL-17 expression assay. Results: TR-FRET binding assay and SPR biosensor assay revealed rapid, reversible, and high affinity binding of TAK-828F to RORγt. The cofactor recruitment assay showed that TAK-828F inhibited the recruitment of steroid receptor coactivator-1 to RORγt. Furthermore, TAK-828F inhibited the transcriptional activity of human and mouse RORγt with selectivity against human RORα and RORβ. TAK-828F also suppressed IL-17 production in Jurkat cells, overexpressing human RORγt. Conclusion: These favorable properties will be of advantage in the evaluation of TAK-828F in clinical studies for inflammatory diseases. Furthermore, these findings demonstrate that TAK-828F could serve as a pharmacological tool for further studies of RORγt and inflammatory diseases.


Bioorganic & Medicinal Chemistry | 2018

Structure-based design, synthesis, and biological evaluation of imidazo[1,2- b ]pyridazine-based p38 MAP kinase inhibitors

Akira Kaieda; Masashi Takahashi; Takafumi Takai; Masayuki Goto; Takahiro Miyazaki; Yuri Hori; Satoko Unno; Tomohiro Kawamoto; Toshimasa Tanaka; Sachiko Itono; Terufumi Takagi; Teruki Hamada; Mikio Shirasaki; Kengo Okada; Gyorgy Snell; Ken Bragstad; Bi-Ching Sang; Osamu Uchikawa; Seiji Miwatashi

We identified novel potent inhibitors of p38 MAP kinase using structure-based design strategy. X-ray crystallography showed that when p38 MAP kinase is complexed with TAK-715 (1) in a co-crystal structure, Phe169 adopts two conformations, where one interacts with 1 and the other shows no interaction with 1. Our structure-based design strategy shows that these two conformations converge into one via enhanced protein-ligand hydrophobic interactions. According to the strategy, we focused on scaffold transformation to identify imidazo[1,2-b]pyridazine derivatives as potent inhibitors of p38 MAP kinase. Among the herein described and evaluated compounds, N-oxide 16 exhibited potent inhibition of p38 MAP kinase and LPS-induced TNF-α production in human monocytic THP-1 cells, and significant in vivo efficacy in rat collagen-induced arthritis models. In this article, we report the discovery of potent, selective and orally bioavailable imidazo[1,2-b]pyridazine-based p38 MAP kinase inhibitors with pyridine N-oxide group.


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.


Assay and Drug Development Technologies | 2018

Cholesterol Unbound RORγt Protein Enables a Sensitive Inverse Agonist Screening

Ryokichi Koyama; Yasunori Fukuda; Yusuke Kamada; Hideyuki Nakagawa; Darbi Witmer; Geza Ambrus-Aikelin; Bi-Ching Sang; Masaharu Nakayama; Hidehisa Iwata

The retinoic acid-related orphan receptor gamma T (RORγt) plays an important role in Th17 cell proliferation and functionality. Thus, RORγt inverse agonists are thought to be potent therapeutic agents for Th17-mediated autoimmune diseases, such as rheumatoid arthritis, asthma, inflammatory bowel disease, and psoriasis. Although RORγt has constitutive activity, it is recognized that the receptor is physiologically regulated by various cholesterol derivatives. In this study, we sought to identify RORγt inverse agonists through a high-throughput screening campaign. To this end, we compared an apo-RORγt protein from Escherichia coli and a cholesterol-bound RORγt protein from insect cells. The IC50 of the known RORγt inverse agonist TO901317 was significantly lower for the apoprotein than for the cholesterol-bound RORγt. Through high-throughput screening using a fluorescence-based cholesterol binding assay with the apoprotein, we identified compound 1 as a novel cholesterol-competitive RORγt inverse agonist. Compound 1 inhibited the RORγt-TopFluor cholesterol interaction, coactivator recruitment, and transcriptional activity of RORγt. Cell-based reporter gene assay demonstrated that compound 1 showed higher potency by lipid depletion treatment. Collectively, our findings indicate that eliminating cholesterol from the RORγt protein is suitable for sensitive high-throughput screening to identify RORγt inverse agonists.


Journal of Molecular Biology | 2016

Structure of Human GIVD Cytosolic Phospholipase A2 Reveals Insights into Substrate Recognition

Hui Wang; Michael G. Klein; Gyorgy Snell; Weston Lane; Hua Zou; Irena Levin; Ke Li; Bi-Ching Sang

Cytosolic phospholipases A2 (cPLA2s) consist of a family of calcium-sensitive enzymes that function to generate lipid second messengers through hydrolysis of membrane-associated glycerophospholipids. The GIVD cPLA2 (cPLA2δ) is a potential drug target for developing a selective therapeutic agent for the treatment of psoriasis. Here, we present two X-ray structures of human cPLA2δ, capturing an apo state, and in complex with a substrate-like inhibitor. Comparison of the apo and inhibitor-bound structures reveals conformational changes in a flexible cap that allows the substrate to access the relatively buried active site, providing new insight into the mechanism for substrate recognition. The cPLA2δ structure reveals an unexpected second C2 domain that was previously unrecognized from sequence alignments, placing cPLA2δ into the class of membrane-associated proteins that contain a tandem pair of C2 domains. Furthermore, our structures elucidate novel inter-domain interactions and define three potential calcium-binding sites that are likely important for regulation and activation of enzymatic activity. These findings provide novel insights into the molecular mechanisms governing cPLA2s function in signal transduction.

Collaboration


Dive into the Bi-Ching Sang's collaboration.

Top Co-Authors

Avatar

Hideyuki Nakagawa

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Atsuko Ochida

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Gyorgy Snell

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Junya Shirai

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Masaharu Nakayama

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Mitsunori Kono

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Robert J. Skene

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Satoshi Yamamoto

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Isaac D. Hoffman

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Kazuko Yonemori

Takeda Pharmaceutical Company

View shared research outputs
Researchain Logo
Decentralizing Knowledge