Network


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

Hotspot


Dive into the research topics where Yuchi Ma is active.

Publication


Featured researches published by Yuchi Ma.


Journal of Medicinal Chemistry | 2015

Design and Optimization of a Series of 1-Sulfonylpyrazolo[4,3-b]pyridines as Selective c-Met Inhibitors

Yuchi Ma; Guangqiang Sun; Danqi Chen; Xia Peng; Yue-Lei Chen; Yi Su; Yinchun Ji; Jin Liang; Xin Wang; Lin Chen; Jian Ding; Bing Xiong; Jing Ai; Meiyu Geng; Jingkang Shen

c-Met has emerged as an attractive target for targeted cancer therapy because of its abnormal activation in many cancer cells. To identify high potent and selective c-Met inhibitors, we started with profiling the potency and in vitro metabolic stability of a reported hit 7. By rational design, a novel sulfonylpyrazolo[4,3-b]pyridine 9 with improved DMPK properties was discovered. Further elaboration of π-π stacking interactions and solvent accessible polar moieties led to a series of highly potent and selective type I c-Met inhibitors. On the basis of in vitro and in vivo pharmacological and pharmacokinetics studies, compound 46 was selected as a preclinical candidate for further anticancer drug development.


Acta Pharmacologica Sinica | 2013

Energetic factors determining the binding of type I inhibitors to c-Met kinase: experimental studies and quantum mechanical calculations

Yuchi Ma; Jing Ai; Danqi Chen; Dongmei Zhao; Xin Wang; Yue-Lei Chen; Meiyu Geng; Bing Xiong; Mao-sheng Cheng; Jingkang Shen

Aim:To decipher the molecular interactions between c-Met and its type I inhibitors and to facilitate the design of novel c-Met inhibitors.Methods:Based on the prototype model inhibitor 1, four ligands with subtle differences in the fused aromatic rings were synthesized. Quantum chemistry was employed to calculate the binding free energy for each ligand. Symmetry-adapted perturbation theory (SAPT) was used to decompose the binding energy into several fundamental forces to elucidate the determinant factors.Results:Binding free energies calculated from quantum chemistry were correlated well with experimental data. SAPT calculations showed that the predominant driving force for binding was derived from a sandwich π–π interaction with Tyr-1230. Arg-1208 was the differentiating factor, interacting with the 6-position of the fused aromatic ring system through the backbone carbonyl with a force pattern similar to hydrogen bonding. Therefore, a hydrogen atom must be attached at the 6-position, and changing the carbon atom to nitrogen caused unfavorable electrostatic interactions.Conclusion:The theoretical studies have elucidated the determinant factors involved in the binding of type I inhibitors to c-Met.


ChemMedChem | 2012

Discovery of 3H-Imidazo[4,5-b]pyridines as Potent c-Met Kinase Inhibitors: Design, Synthesis, and Biological Evaluation

Danqi Chen; Ying Wang; Yuchi Ma; Bing Xiong; Jing Ai; Yi Chen; Meiyu Geng; Jingkang Shen

To identify novel c‐Met inhibitors, sequences and crystal structures of the human kinome were analyzed to find interesting hinge binders that have been underexplored within the tyrosine kinase subfamily. Through this study, the imidazolopyridine ring was selected as a novel c‐Met hinge‐binding inhibitor scaffold. A series of derivatives was prepared, and the structure–activity relationships were studied. Among these, one compound in particular showed excellent activities in enzymatic and cellular assays, good in vitro metabolic stability, and favorable pharmacokinetic parameters. When administered orally, the compound inhibited tumor growth in an NIH‐3T3/TPR‐Met xenograft model and did not show adverse effects on body weight. The present work not only conceptually demonstrates a new route for designing novel kinase inhibitors by using known structural information of ligand–hinge interactions but also provides a series of imidazolopyridine derivatives as potent c‐Met inhibitors.


RSC Advances | 2014

Condensed Fukui function predicts innate C–H radical functionalization sites on multi-nitrogen containing fused arenes

Yuchi Ma; Jin Liang; Dongmei Zhao; Yue-Lei Chen; Jingkang Shen; Bing Xiong

The condensed Fukui function could be correlated with the reported innate C–H radical functionalization sites on some heterocycles. This computational method was further extended to predict the innate C–H functionalization sites on multi-nitrogen containing fused arenes, and the calculated results were validated by experimental outcomes.


Molecules | 2017

Discovery and Biological Evaluation of a Series of Pyrrolo[2,3-b]pyrazines as Novel FGFR Inhibitors

Yan Zhang; Hongchun Liu; Zhen Zhang; Ruifeng Wang; Tongchao Liu; Chaoyun Wang; Yuchi Ma; Jing Ai; Dongmei Zhao; Jingkang Shen; Bing Xiong

Abnormality of fibroblast growth factor receptor (FGFR)-mediated signaling pathways were frequently found in various human malignancies, making FGFRs hot targets for cancer treatment. To address the consistent need for a new chemotype of FGFR inhibitors, here, we started with a hit structure identified from our internal hepatocyte growth factor receptor (also called c-Met) inhibitor project, and conducted a chemical optimization. After exploring three parts of the hit compound, we finally discovered a new series of pyrrolo[2,3-b]pyrazine FGFR inhibitors, which contain a novel scaffold and unique molecular shape. We believe that our findings can help others to further develop selective FGFR inhibitors.


Molecules | 2016

Design, Synthesis and Biological Evaluation of 6-(2,6-Dichloro-3,5-dimethoxyphenyl)- 4-substituted-1H-indazoles as Potent Fibroblast Growth Factor Receptor Inhibitors

Zhen Zhang; Dongmei Zhao; Yang Dai; Mao-sheng Cheng; Meiyu Geng; Jingkang Shen; Yuchi Ma; Jing Ai; Bing Xiong

Tyrosine kinase fibroblast growth factor receptor (FGFR), which is aberrant in various cancer types, is a promising target for cancer therapy. Here we reported the design, synthesis, and biological evaluation of a new series of 6-(2,6-dichloro-3,5-dimethoxyphenyl)-4-substituted-1H-indazole derivatives as potent FGFR inhibitors. The compound 6-(2,6-dichloro-3,5-dimethoxyphenyl)-N-phenyl-1H-indazole-4-carboxamide (10a) was identified as a potent FGFR1 inhibitor, with good enzymatic inhibition. Further structure-based optimization revealed that 6-(2,6-dichloro-3,5-dimethoxyphenyl)-N-(3-(4-methylpiperazin-1-yl)phenyl)-1H-indazole-4-carboxamide (13a) is the most potent FGFR1 inhibitor in this series, with an enzyme inhibitory activity IC50 value of about 30.2 nM.


Acta Pharmacologica Sinica | 2016

Discovery of a new series of imidazo[1,2-a]pyridine compounds as selective c-Met inhibitors

Tongchao Liu; Xia Peng; Yuchi Ma; Yinchun Ji; Danqi Chen; Mingyue Zheng; Dongmei Zhao; Mao-sheng Cheng; Meiyu Geng; Jingkang Shen; Jing Ai; Bing Xiong

Aim:Aberrant c-Met activation plays a critical role in cancer formation, progression and dissemination, as well as in development of resistance to anticancer drugs. Therefore, c-Met has emerged as an attractive target for cancer therapy. The aim of this study was to develop new c-Met inhibitors and elaborate the structure-activity relationships of identified inhibitors.Methods:Based on the predicted binding modes of Compounds 5 and 14 in docking studies, a new series of c-Met inhibitor-harboring 3-((1H-pyrrolo[3,2-c]pyridin-1-yl)sulfonyl)imidazo[1,2-a]pyridine scaffolds was discovered. Potent inhibitors were identified through extensive optimizations combined with enzymatic and cellular assays. A promising compound was further investigated in regard to its selectivity, its effects on c-Met signaling, cell proliferation and cell scattering in vitro.Results:The most potent Compound 31 inhibited c-Met kinase activity with an IC50 value of 12.8 nmol/L, which was >78-fold higher than those of a panel of 16 different tyrosine kinases. Compound 31 (8, 40, 200 nmol/L) dose-dependently inhibited the phosphorylation of c-Met and its key downstream Akt and ERK signaling cascades in c-Met aberrant human EBC-1 cancer cells. In 12 human cancer cell lines harboring different background levels of c-Met expression/activation, Compound 31 potently inhibited c-Met-driven cell proliferation. Furthermore, Compound 31 dose-dependently impaired c-Met-mediated cell scattering of MDCK cells.Conclusion:This series of c-Met inhibitors is a promising lead for development of novel anticancer drugs.


Molecular Cancer Therapeutics | 2017

Preclinical Evaluation of SCC244 (Glumetinib), a Novel, Potent, and Highly Selective Inhibitor of c-Met in MET-dependent Cancer Models

Jing Ai; Yi Chen; Xia Peng; Yinchun Ji; Yong Xi; Yanyan Shen; Xinying Yang; Yi Su; Yiming Sun; Yinglei Gao; Yuchi Ma; Bing Xiong; Jingkang Shen; Jian Ding; Meiyu Geng

Because the receptor tyrosine kinase c-Met plays a critical role in tumor growth, metastasis, tumor angiogenesis, and drug resistance, the c-Met axis represents an attractive therapeutic target. Herein, we report the first preclinical characterization of SCC244, a novel, potent, and highly selective inhibitor of c-Met kinase. SCC244 showed subnanomolar potency against c-Met kinase activity and high selectivity versus 312 other tested protein kinases, making it one of the most selective c-Met inhibitors described to date. Moreover, this inhibitor profoundly and specifically inhibits c-Met signal transduction and thereby suppresses the c-Met–dependent neoplastic phenotype of tumor and endothelial cells. In xenografts of human tumor cell lines or non–small cell lung cancer and hepatocellular carcinoma patient-derived tumor tissue driven by MET aberration, SCC244 administration exhibits robust antitumor activity at the well-tolerated doses. In addition, the in vivo antitumor activity of SCC244 involves the inhibition of c-Met downstream signaling via a mechanism of combined antiproliferation and antiangiogenic effects. The results of the current study provide a strong foundation for the clinical investigation of SCC244 in patients with tumors harboring c-Met pathway alterations. Mol Cancer Ther; 17(4); 751–62. ©2017 AACR.


Molecules | 2018

Structure-Based Discovery of a Series of 5H-Pyrrolo[2,3-b]pyrazine FGFR Kinase Inhibitors

Alan Jiang; Qiufeng Liu; Ruifeng Wang; Peng Wei; Yang Dai; Xin Wang; Yechun Xu; Yuchi Ma; Jing Ai; Jingkang Shen; Jian Ding; Bing Xiong

Fibroblast growth factor receptors (FGFRs), a subfamily of receptor tyrosine kinases, are aberrant in various cancer types, and considered to be promising targets for cancer therapy. We started with a weak-active compound that was identified from our internal hepatocyte growth factor receptor (also called c-Met) inhibitor project, and optimized it with the guidance of a co-crystal structure of compound 8 with FGFR1. Through rational design, synthesis, and the biological evaluation of a series of 5H-pyrrolo[2,3-b]pyrazine derivatives, we discovered several potent FGFR kinase inhibitors. Among them, compound 13 displayed high selectivity and favorable metabolic properties, demonstrating a promising lead for further development.


Tetrahedron Letters | 2016

Formation of 1,3-diazocine by palladium catalyzed C–H arylation

Le Tang; Jing Ren; Yuchi Ma; Xin Wang; Lin Chen; Jingkang Shen; Yue-Lei Chen; Bing Xiong

Collaboration


Dive into the Yuchi Ma's collaboration.

Top Co-Authors

Avatar

Bing Xiong

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Jingkang Shen

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Jing Ai

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Meiyu Geng

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Danqi Chen

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Dongmei Zhao

Shenyang Pharmaceutical University

View shared research outputs
Top Co-Authors

Avatar

Xin Wang

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Yue-Lei Chen

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Jian Ding

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Xia Peng

Chinese Academy of Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge