Network


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

Hotspot


Dive into the research topics where Xinying Yang is active.

Publication


Featured researches published by Xinying Yang.


European Journal of Medicinal Chemistry | 2014

Discovery of potent N-(isoxazol-5-yl)amides as HSP90 inhibitors.

Danqi Chen; Aijun Shen; Jian Li; Feng Shi; Wuyan Chen; Jing Ren; Hongchun Liu; Yechun Xu; Xin Wang; Xinying Yang; Yiming Sun; Min Yang; Jianhua He; Yueqin Wang; Liping Zhang; Min Huang; Meiyu Geng; Bing Xiong; Jingkang Shen

HSP90 is ubiquitously overexpressed in a broad spectrum of human cancers and has been recognized as an attractive target for cancer treatment. Here, we described the fragment screening, synthesis and structure-activity relationship studies of small molecule inhibitors with 4,5-diarylisoxazole scaffold targeting HSP90. Among them, the compound N-(3-(2,4-dihydroxy-5-isopropylphenyl)-4-(4-((4-morpholinopiperidin-1-yl)methyl)phenyl)isoxazol-5-yl)cyclopropanecarboxamide (108) showed high affinity for binding to HSP90 (FP binding assay, IC50 = 0.030 μM) and inhibited the proliferation of various human cancer cell lines with averaging GI50 about 88 nM. Compound 108 exhibited its functional inhibition of HSP90 by depleting key signaling pathways and concomitantly elevating of HSP70 and HSP27 in U-87MG cells. Further in vivo studies showed that compound 108 strongly suppressed the tumor growth of human glioblastoma xenograft model U-87MG with T/C = 18.35% at 50 mg/kg q3w/2.5w. Moreover, compound 108 also exhibited good pharmacokinetic properties. Together, our study implicates that compound 108 is a promising candidate of HSP90 inhibitor and is currently advanced to preclinical study.


Cancer Cell | 2016

Feedback Activation of Leukemia Inhibitory Factor Receptor Limits Response to Histone Deacetylase Inhibitors in Breast Cancer

Hanlin Zeng; Jia Qu; Nan Jin; Jun Xu; Chenchu Lin; Yi Chen; Xinying Yang; Xiang He; Shuai Tang; Xiaojing Lan; Xiaotong Yang; Ziqi Chen; Min Huang; Jian Ding; Meiyu Geng

Histone deacetylase (HDAC) inhibitors have demonstrated clinical benefits in subtypes of hematological malignancies. However, the efficacy of HDAC inhibitors in solid tumors remains uncertain. This study takes breast cancer as a model to understand mechanisms accounting for limited response of HDAC inhibitors in solid tumors and to seek combination solutions. We discover that feedback activation of leukemia inhibitory factor receptor (LIFR) signaling in breast cancer limits the response to HDAC inhibition. Mechanistically, HDAC inhibition increases histone acetylation at the LIFR gene promoter, which recruits bromodomain protein BRD4, upregulates LIFR expression, and activates JAK1-STAT3 signaling. Importantly, JAK1 or BRD4 inhibition sensitizes breast cancer to HDAC inhibitors, implicating combination inhibition of HDAC with JAK1 or BRD4 as potential therapies for breast cancer.


Journal of Medicinal Chemistry | 2016

Discovery of 3-(5'-Substituted)-Benzimidazole-5-(1-(3,5-dichloropyridin-4-yl)ethoxy)-1H-indazoles as Potent Fibroblast Growth Factor Receptor Inhibitors: Design, Synthesis, and Biological Evaluation.

Wei Yan; Xinyi Wang; Yang Dai; Bin Zhao; Xinying Yang; Jun Fan; Yinglei Gao; Fanwang Meng; Yuming Wang; Cheng Luo; Jing Ai; Meiyu Geng; Wenhu Duan

Fibroblast growth factor receptor (FGFR) represents an attractive oncology target for cancer therapy in view of its critical role in promoting cancer formation and progression, as well as causing resistance to approved therapies. In this article, we describe the identification of the potent pan-FGFR inhibitor (R)-21c (FGFR1-4 IC50 values of 0.9, 2.0, 2.0, and 6.1 nM, respectively). Compound (R)-21c exhibited excellent in vitro inhibitory activity against a panel of FGFR-amplified cell lines. Western blot analysis demonstrated that (R)-21c suppressed FGF/FGFR and downstream signaling pathways at nanomolar concentrations. Moreover, (R)-21c provided nearly complete inhibition of tumor growth (96.9% TGI) in NCI-H1581 (FGFR1-amplified) xenograft mice model at the dose of 10 mg/kg/qd via oral administration.


Cancer Research | 2015

c-Myc alterations confer therapeutic response and acquired resistance to c-Met inhibitors in MET-addicted cancers.

Aijun Shen; Lu Wang; Min Huang; Jingya Sun; Yi Chen; Yanyan Shen; Xinying Yang; Xin Wang; Jian Ding; Meiyu Geng

Use of kinase inhibitors in cancer therapy leads invariably to acquired resistance stemming from kinase reprogramming. To overcome the dynamic nature of kinase adaptation, we asked whether a signal-integrating downstream effector might exist that provides a more applicable therapeutic target. In this study, we reported that the transcriptional factor c-Myc functions as a downstream effector to dictate the therapeutic response to c-Met inhibitors in c-Met-addicted cancer and derived resistance. Dissociation of c-Myc from c-Met control, likely overtaken by a variety of reprogrammed kinases, led to acquisition of drug resistance. Notably, c-Myc blockade by RNA interference or pharmacologic inhibition circumvented the acquired resistance to c-Met inhibition. Combining c-Myc blockade and c-Met inhibition in MET-amplified patient-derived xenograft mouse models heightened therapeutic activity. Our findings offer a preclinical proof of concept for the application of c-Myc-blocking agents as a tactic to thwart resistance to kinase inhibitors.


Oncogene | 2017

Phosphoglycerate mutase 1 promotes cancer cell migration independent of its metabolic activity

Dadong Zhang; Nan Jin; Wenyi Sun; X Li; B Liu; Zuoquan Xie; Jia Qu; Jun Xu; Xinying Yang; Yi Su; Shuai Tang; H Han; D Chen; Jian Ding; Minjia Tan; Min Huang; Meiyu Geng

Phosphoglycerate mutase 1 (PGAM1) is a glycolytic enzyme that coordinates glycolysis and biosynthesis to promote cancer growth via its metabolic activity. Here, we report the discovery of a non-metabolic function of PGAM1 in promoting cancer metastasis. A proteomic study identified α-smooth muscle actin (ACTA2) as a PGAM1-associated protein. PGAM1 modulated actin filaments assembly, cell motility and cancer cell migration via directly interacting with ACTA2, which was independent of its metabolic activity. The enzymatically inactive H186R mutant retained its association with ACTA2, whereas 201–210 amino acids deleted PGAM1 mutant lost the interaction with ACTA2 regardless of intact metabolic activity. Importantly, PGAM1 knockdown decreased metastatic potential of breast cancer cells in vivo and PGAM1 and ACTA2 were jointly associated with the prognosis of breast cancer patients. Together, this study provided the first evidence revealing a non-metabolic function of PGAM1 in promoting cell migration, and gained new insights into the role of PGAM1 in cancer progression.


Oncotarget | 2017

Novel PARP1/2 inhibitor mefuparib hydrochloride elicits potent in vitro and in vivo anticancer activity, characteristic of high tissue distribution.

Jinxue He; Meng Wang; Xiajuan Huan; Chuanhuizi Chen; Shanshan Song; Ying-Qing Wang; Xue-mei Liao; Cun Tan; Qian He; Linjiang Tong; Yu-Ting Wang; Xiao-hua Li; Yi Su; Yanyan Shen; Yiming Sun; Xinying Yang; Yi Chen; Zhiwei Gao; Xiao-Yan Chen; Bing Xiong; Xiu-Lian Lu; Jian Ding; Chunhao Yang; Ze-Hong Miao

The approval of poly(ADP-ribose) polymerase (PARP) inhibitor AZD2281 in 2014 marked the successful establishment of the therapeutic strategy targeting homologous recombination repair defects of cancers in the clinic. However, AZD2281 has poor water solubility, low tissue distribution and relatively weak in vivo anticancer activity, which appears to become limiting factors for its clinical use. In this study, we found that mefuparib hydrochloride (MPH) was a potent PARP inhibitor, possessing prominent in vitro and in vivo anticancer activity. Notably, MPH displayed high water solubility (> 35 mg/ml) and potent PARP1/2 inhibition in a substrate-competitive manner. It reduced poly(ADP-ribose) (PAR) formation, enhanced γH2AX levels, induced G2/M arrest and subsequent apoptosis in homologous recombination repair (HR)-deficient cells. Proof-of-concept studies confirmed the MPH-caused synthetic lethality. MPH showed potent in vitro and in vivo proliferation and growth inhibition against HR-deficient cancer cells and synergistic sensitization of HR-proficient xenografts to the anticancer drug temozolomide. A good relationship between the anticancer activity and the PARP inhibition of MPH suggested that PAR formation and γH2AX accumulation could serve as its pharmacodynamic biomarkers. Its high bioavailability (40%~100%) and high tissue distribution in both monkeys and rats were its most important pharmacokinetic features. Its average concentrations were 33-fold higher in the tissues than in the plasma in rats. Our work supports the further clinical development of MPH as a novel PARP1/2 inhibitor for cancer therapy.


Hepatology | 2017

Polymeric immunoglobulin receptor promotes tumor growth in hepatocellular carcinoma

Xihua Yue; Jing Ai; Yang Xu; Yi Chen; Min Huang; Xinying Yang; Bo Hu; Hao-tian Zhang; Chang-xi He; Xin-Rong Yang; Wei-Guo Tang; Xia Peng; Liwei Dong; Wang H; Jia Fan; Jian Ding; Meiyu Geng

Deregulation of the immune system is believed to contribute to cancer malignancy, which has led to recent therapeutic breakthroughs facilitating antitumor immunity. In a malignant setting, immunoglobulin receptors, which are fundamental components of the human immune system, fulfill paradoxical roles in cancer pathogenesis. This study describes a previously unrecognized pro‐oncogenic function of polymeric immunoglobulin receptor (pIgR) in the promotion of cell transformation and proliferation. Mechanistically, pIgR overexpression is associated with YES proto‐oncogene 1, Src family tyrosine kinase (Yes) activation, which is required for pIgR‐induced oncogenic growth. Specifically, pIgR activates the Yes‐DNAX‐activating protein of 12 kDa‐spleen tyrosine kinase‐Rac1/CDC42‐MEK (extracellular signal‐regulated kinase kinase)/ERK (extracellular signal‐regulated kinase) cascade in an immunoreceptor tyrosine‐based activating motif (ITAM)‐dependent manner to promote cell transformation and tumor growth, although pIgR itself does not contain an ITAM sequence. Additionally, the combination of pIgR and phosphorylated Yes (p‐Yes) levels serves as a prognostic biomarker for hepatitis B surface antigen–positive and early‐stage hepatocellular carcinoma (HCC) patients. Moreover, pharmacological targeting of MEK/ERK or Yes represents a therapeutic option for the subgroup of patients with pIgR/p‐Yes–positive HCC based on our results with both cancer cell‐line–based xenografts and primary patient‐derived xenografts. Conclusion: Our findings reveal the molecular mechanism by which pIgR promotes cancer malignancy, suggest the clinical potential of targeting this pathway in HCC, and provide new insight into the oncogenic role of immunoglobulin receptors. (Hepatology 2017;65:1948‐1962).


Acta Pharmacologica Sinica | 2017

Combining 53BP1 with BRCA1 as a biomarker to predict the sensitivity of poly(ADP-ribose) polymerase (PARP) inhibitors

Zhong-min Yang; Xue-mei Liao; Yi Chen; Yanyan Shen; Xinying Yang; Yi Su; Yiming Sun; Yinglei Gao; Jian Ding; Ao Zhang; Jinxue He; Ze-Hong Miao

Over half of patients with BRCA1-deficient cancers do not respond to treatment with poly(ADP-ribose) polymerase (PARP) inhibitors. In this study, we report that a combination of 53BP1 and BRCA1 may serve as a biomarker of PARP inhibitor sensitivity. Based on the mRNA levels of four homologous recombination repair (HR) genes and PARP inhibitor sensitivity, we selected BRCA1-deficient MDA-MB-436 cells to conduct RNA interference. Reducing expression of 53BP1, but not the other three HR genes, was found to lower simmiparib sensitivity. Additionally, we generated 53BP1−/−/BRCA1−/− clonal variants by the transcription activator-like effector nuclease (TALEN) technique and found that depleting 53BP1 impaired PARP inhibitor sensitivity with a 36.7-fold increase in their IC50 values. Consistent with its effect on PARP inhibitor sensitivity, 53BP1 loss alleviated cell cycle arrest and apoptosis and partially restored HR function. Importantly, 53BP1 depletion dramatically reduced the ability of PARP inhibitors to suppress tumor growth in vivo. The inhibition rate of simmiparib was 74.16% for BRCA1-deficient MDA-MB-436 xenografts, but only 7.79% for 53BP1/BRCA1-deficient xenografts. Re-expressing 53BP1 in the dual-deficient cells restored PARP inhibitor sensitivity and the levels of HR regulators. Considering that at least 10% of BRCA1-deficient breast and ovarian cancers have reduced expression of 53BP1, using a combination of 53BP1 with BRCA1 as a biomarker for patient selection should reduce the number of patients undergoing futile treatment with PARP inhibitors.


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.


Theranostics | 2017

Integration of Receptor Tyrosine Kinases Determines Sensitivity to PI3Kα-selective Inhibitors in Breast Cancer

Yi-chao Xu; Xiang Wang; Yi Chen; Si-meng Chen; Xinying Yang; Yiming Sun; Meiyu Geng; Jian Ding; Linghua Meng

PI3Kα-selective inhibitor BYL719 is currently in phase II/III clinical trial for the treatment of breast cancer, but highly variable response has been observed among patients. We sought to discover predictive biomarker for the efficacy of BYL719 by dissecting the proliferative signaling pathway mediated by PI3K in breast cancer. BYL719 concurrently inhibited the phosphorylation of AKT and ERK in PIK3CA-mutated human breast cancer cells. PI3K-regulated ERK phosphorylation was independent of canonical PDK1/AKT/mTOR pathway, while it was associated with RAF/MEK. Hyper-activation of EGFR or RAS abrogated inhibition of ERK phosphorylation by BYL719. Furthermore, hyper-activation of receptor tyrosine kinases (RTKs) including EGFR, c-MET, FGFR and HER3 but not IGF-1R restored ERK phosphorylation and cell viability suppressed by BYL719, suggesting the discriminative functions of RTKs in cell signaling and proliferation. By profiling 22 breast cancer cell lines, we found that BYL719 was more potent in cell lines where phosphorylation of both AKT and ERK was attenuated than those where only AKT phosphorylation was inhibited. The potency of BYL719 was further found to be significantly correlated with the expression profile of RTKs in breast cancer cells. Specifically, overexpression of EGFR, c-MET and/or FGFR1 forecasted resistance, while overexpression of IGF-1R and/or HER2 predicted sensitivity to BYL719 in breast cancer cells. Similar correlation between BYL719 efficacy and expression profile of RTKs was found in patient-derived xenograft models of breast cancer. Thus, inhibition of ERK phosphorylation by PI3Kα inhibitor BYL719 contributes to its antitumor efficacy and is determined by the converged signaling from RTKs. The expression profile of RTKs in breast cancer tissue could be potentially developed as a predictive biomarker for the efficacy of PI3Kα inhibitors.

Collaboration


Dive into the Xinying Yang's collaboration.

Top Co-Authors

Avatar

Jian Ding

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

Yi Chen

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Min Huang

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Yiming Sun

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Yanyan Shen

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Yi Su

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

Yinglei Gao

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Bing Xiong

Chinese Academy of Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge