Network


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

Hotspot


Dive into the research topics where Yujia Dai is active.

Publication


Featured researches published by Yujia Dai.


Molecular Cancer Therapeutics | 2006

Preclinical activity of ABT-869, a multitargeted receptor tyrosine kinase inhibitor

Daniel H. Albert; Paul Tapang; Terrance J. Magoc; Lori J. Pease; David R. Reuter; Ru-Qi Wei; Junling Li; Jun Guo; Peter F. Bousquet; Nayereh S. Ghoreishi-Haack; Baole Wang; Gail T. Bukofzer; Yi-Chun Wang; Jason Stavropoulos; Kresna Hartandi; Amanda Niquette; Nirupama B. Soni; Eric F. Johnson; J. Owen McCall; Jennifer J. Bouska; Yanping Luo; Cherrie K. Donawho; Yujia Dai; Patrick A. Marcotte; Keith B. Glaser; Michael R. Michaelides; Steven K. Davidsen

ABT-869 is a structurally novel, receptor tyrosine kinase (RTK) inhibitor that is a potent inhibitor of members of the vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF) receptor families (e.g., KDR IC50 = 4 nmol/L) but has much less activity (IC50s > 1 μmol/L) against unrelated RTKs, soluble tyrosine kinases, or serine/threonine kinases. The inhibition profile of ABT-869 is evident in cellular assays of RTK phosphorylation (IC50 = 2, 4, and 7 nmol/L for PDGFR-β, KDR, and CSF-1R, respectively) and VEGF-stimulated proliferation (IC50 = 0.2 nmol/L for human endothelial cells). ABT-869 is not a general antiproliferative agent because, in most cancer cells, >1,000-fold higher concentrations of ABT-869 are required for inhibition of proliferation. However, ABT-869 exhibits potent antiproliferative and apoptotic effects on cancer cells whose proliferation is dependent on mutant kinases, such as FLT3. In vivo ABT-869 is effective orally in the mechanism-based murine models of VEGF-induced uterine edema (ED50 = 0.5 mg/kg) and corneal angiogenesis (>50% inhibition, 15 mg/kg). In tumor growth studies, ABT-869 exhibits efficacy in human fibrosarcoma and breast, colon, and small cell lung carcinoma xenograft models (ED50 = 1.5–5 mg/kg, twice daily) and is also effective (>50% inhibition) in orthotopic breast and glioma models. Reduction in tumor size and tumor regression was observed in epidermoid carcinoma and leukemia xenograft models, respectively. In combination, ABT-869 produced at least additive effects when given with cytotoxic therapies. Based on pharmacokinetic analysis from tumor growth studies, efficacy correlated more strongly with time over a threshold value (cellular KDR IC50 corrected for plasma protein binding = 0.08 μg/mL, ≥7 hours) than with plasma area under the curve or Cmax. These results support clinical assessment of ABT-869 as a therapeutic agent for cancer. [Mol Cancer Ther 2006;5(4):995–1006]


Molecular Cancer Therapeutics | 2006

Inhibition of phosphorylation of the colony-stimulating factor-1 receptor (c-Fms) tyrosine kinase in transfected cells by ABT-869 and other tyrosine kinase inhibitors

Jun Guo; Patrick A. Marcotte; J. Owen McCall; Yujia Dai; Lori J. Pease; Michael R. Michaelides; Steven K. Davidsen; Keith B. Glaser

The properties of several multitargeted receptor tyrosine kinase inhibitors have been studied for their inhibition of colony-stimulating factor-1 receptor (CSF-1R) signaling. A structurally novel, multitargeted tyrosine kinase inhibitor (ABT-869), imatinib (STI571), and four compounds currently in clinical development (AG013736, BAY 43-9006, CHIR258, and SU11248) were tested for inhibition of CSF-1R signaling in both the enzymatic and cellular assays. ABT-869 showed potent CSF-1R inhibition in both the enzyme and cell-based assays (IC50s < 20 nmol/L). In contrast to a previous report, we have found that imatinib has activity against human CSF-1R in both assays at submicromolar concentrations. In enzyme assays, we have found that the inhibition of CSF-1R by both ABT-869 and imatinib are competitive with ATP, with Ki values of 3 and 120 nmol/L, respectively. SU11248 is a potent inhibitor of CSF-1R in the enzyme assay (IC50 = 7 nmol/L) and inhibits receptor phosphorylation in the cellular assay (IC50 = 61 nmol/L). AG013736 was also a potent inhibitor of CSF-1R in both assays (enzyme, IC50 = 16 nmol/L; cellular, IC50 = 21 nmol/L), whereas BAY 43-9006 is less potent in the enzyme assay (IC50 = 107 nmol/L) than in the cellular system (IC50 = 20 nmol/L). In contrast, we found that CHIR258 had less activity in the cellular assay (IC50 = 535 nmol/L) relative to its enzymatic potency (IC50 = 26 nmol/L). These results show the use of a cell-based assay to confirm the inhibitory activity of lead compounds and drug candidates, such as ABT-869, against the CSF-1R protein in situ. [Mol Cancer Ther 2006;5(4):1007–13]


Bioorganic & Medicinal Chemistry Letters | 2003

Indole amide hydroxamic acids as potent inhibitors of histone deacetylases.

Yujia Dai; Yan Guo; Jun Guo; Lori J. Pease; Junling Li; Patrick A. Marcotte; Keith B. Glaser; Paul Tapang; Daniel H. Albert; Paul L. Richardson; Steven K. Davidsen; Michael R. Michaelides

A series of hydroxamic acid-based HDAC inhibitors with an indole amide residue at the terminus have been synthesized and evaluated. Compounds with a 2-indole amide moiety have been found as the most active inhibitors among the different regioisomers. Introduction of substituents on the indole ring further improved the potency and generated a series of very potent inhibitors with significant antiproliferative activity. A representative compound in the series, 7b, has been found to be orally active in tumor growth inhibition model.


Bioorganic & Medicinal Chemistry Letters | 2011

Discovery of potent and selective thienopyrimidine inhibitors of Aurora kinases.

William J. McClellan; Yujia Dai; Cele Abad-Zapatero; Daniel H. Albert; Jennifer J. Bouska; Keith B. Glaser; Terry Magoc; Patrick A. Marcotte; Donald J. Osterling; Kent D. Stewart; Steven K. Davidsen; Michael R. Michaelides

In an effort to discover Aurora kinase inhibitors, an HTS hit revealed an amide containing pyrrolopyrimidine compound. Replacement of the pyrrolopyrimidine residue with a thienopyrimidine moiety led to a series of potent and selective Aurora inhibitors.


Nature Chemical Biology | 2017

The SUV4-20 inhibitor A-196 verifies a role for epigenetics in genomic integrity

Kenneth D. Bromberg; Taylor R H Mitchell; Anup K. Upadhyay; Clarissa G. Jakob; Manisha A Jhala; Kenneth M. Comess; Loren M. Lasko; Conglei Li; Creighton T. Tuzon; Yujia Dai; Fengling Li; Mohammad S. Eram; Alexander Nuber; Niru B. Soni; Vlasios Manaves; Mikkel A. Algire; Ramzi F. Sweis; Maricel Torrent; Gunnar Schotta; Chaohong Sun; Michael R. Michaelides; Alex R Shoemaker; C.H. Arrowsmith; Peter J. Brown; V. Santhakumar; Alberto Martin; Judd C. Rice; Gary G. Chiang; Masoud Vedadi; Dalia Barsyte-Lovejoy

Protein lysine methyltransferases (PKMTs) regulate diverse physiological processes including transcription and the maintenance of genomic integrity. Genetic studies suggest that the PKMTs SUV420H1 and SUV420H2 facilitate proficient nonhomologous end-joining (NHEJ)-directed DNA repair by catalyzing the di- and trimethylation (me2 and me3, respectively) of lysine 20 on histone 4 (H4K20). Here we report the identification of A-196, a potent and selective inhibitor of SUV420H1 and SUV420H2. Biochemical and co-crystallization analyses demonstrate that A-196 is a substrate-competitive inhibitor of both SUV4-20 enzymes. In cells, A-196 induced a global decrease in H4K20me2 and H4K20me3 and a concomitant increase in H4K20me1. A-196 inhibited 53BP1 foci formation upon ionizing radiation and reduced NHEJ-mediated DNA-break repair but did not affect homology-directed repair. These results demonstrate the role of SUV4-20 enzymatic activity in H4K20 methylation and DNA repair. A-196 represents a first-in-class chemical probe of SUV4-20 to investigate the role of histone methyltransferases in genomic integrity.


Bioorganic & Medicinal Chemistry Letters | 2012

Exploration of diverse hinge-binding scaffolds for selective Aurora kinase inhibitors

Zhiqin Ji; Yujia Dai; Cele Abad-Zapatero; Daniel H. Albert; Jennifer J. Bouska; Keith B. Glaser; Patrick A. Marcotte; Nirupama B. Soni; Terry Magoc; Kent D. Stewart; Ru-Qi Wei; Steve K. Davidsen; Michael R. Michaelides

Four hinge-binding scaffolds have been explored for novel selective Aurora kinase inhibitors. The structure activity relationship, selectivity and pharmacokinetic profiles have been evaluated.


Bioorganic & Medicinal Chemistry Letters | 2018

Discovery and optimization of novel constrained pyrrolopyridone BET family inhibitors

Steven D. Fidanze; Dachun Liu; Robert A. Mantei; Lisa A. Hasvold; John K. Pratt; George S. Sheppard; Le Wang; James H. Holms; Yujia Dai; Ana L. Aguirre; Andrew R. Bogdan; Justin Dietrich; Jasmina Marjanovic; Chang H. Park; Charles W. Hutchins; Xiaoyu Lin; Mai H. Bui; Xiaoli Huang; Denise Wilcox; Leiming Li; Rongqi Wang; Peter Kovar; Terrance J. Magoc; Ganesh Rajaraman; Daniel H. Albert; Yu Shen; Warren M. Kati; Keith F. McDaniel

Novel conformationally constrained BET bromodomain inhibitors have been developed. These inhibitors were optimized in two similar, yet distinct chemical series, the 6-methyl-1H-pyrrolo[2,3-c]pyridin-7(6H)-ones (A) and the 1-methyl-1H-pyrrolo[2,3-c]pyridin-7(6H)-ones (B). Each series demonstrated excellent activity in binding and cellular assays, and lead compounds from each series demonstrated significant efficacy in in vivo tumor xenograft models.


Cancer Research | 2013

Abstract 3450: ABT-348 in combination with inhibition of CDK4/6 highlights a strategy to target RB mutant cells while sparing RB wild-type cells.

Jun Guo; Michael L. Curtin; Zehan Chen; Daniel H. Albert; Paul Tapang; Yujia Dai; Mia-Ha Bui; Peter Kovar; Michael R. Michaelides; Keith B. Glaser; Chris Tse; O. Jameel Shah

Proceedings: AACR 104th Annual Meeting 2013; Apr 6-10, 2013; Washington, DC ABT-348 is a novel, potent and orally bioavailable inhibitor of the Aurora kinases as well as the VEGF and PDGF families of receptor tyrosine kinases. ABT-348 is broadly efficacious preclinically as a single agent against a wide range of tumor types and is currently in Phase I/II clinical trials. Several published studies have supported the potential utility of combining targeted anti-proliferative agents with cytotoxic chemotherapies. Herein, the efficacy of combining the neocytotoxic agent, ABT-348, with CDK4/6 inhibition in the treatment of solid tumor cells has been evaluated. Inhibition of CDK4/6 via siRNA elicits a potent cytostatic response in cells that harbor a wild-type, functional RB. We demonstrate that features of cellular senescence are induced in human tumor cells with siRNAs targeting both CDK4 and CDK6. Dual inhibition of CDK4 and CDK6 is required for antiproliferative activity in most cancer cell lines and this is not accompanied by induction of apoptosis. In RB wild-type cells, CDK4/6 inhibition antagonizes the activity of ABT-348 by inducing G0/G1 arrest. Conversely, CDK4/6 inhibition does not alter the therapeutic response of RB-deficient cells to ABT-348, indicating that the effects of ABT-348 are dependent on cells progressing into mitosis. A preponderance of evidence from previous publications and our studies indicate that CDK4/6 inhibition attenuates the cellular response to cytotoxic chemotherapies in RB wild-type cells providing a potential to expand the therapeutic window for ABT-348. Thus combination with ABT-348 and a CDK4/6 inhibitor may provide an opportunity to selectively target RB mutant cells. Citation Format: Jun Guo, Michael L. Curtin, Zehan Chen, Daniel H. Albert, Paul Tapang, Yujia Dai, Mia-Ha Bui, Peter Kovar, Michael R. Michaelides, Keith B. Glaser, Chris Tse, O. Jameel Shah. ABT-348 in combination with inhibition of CDK4/6 highlights a strategy to target RB mutant cells while sparing RB wild-type cells. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 3450. doi:10.1158/1538-7445.AM2013-3450


Molecular Cancer Therapeutics | 2009

Abstract B14: The broad spectrum antitumor efficacy and tolerability of the RTK inhibitor, linifanib (ABT‐869) alone and in combination with various cytotoxic therapies

Cherrie K. Donawho; Gail Bukofzer; P. Ellis; Debra Montgomery; Luis E. Rodriguez; Yi-Chun Wang; Yujia Dai; Steven K. Davidsen; David J. Frost; Joann P. Palma

The RTKI, linifanib (ABT‐869), is a potent inhibitor of vascular endothelial growth factor (VEGF) as well as platelet‐derived growth factor (PDGF) receptors and is currently in Phase 1 – 2 clinical trials. It exhibits potent inhibition of mutant kinases, such as fms‐like tyrosine kinase 3 (FLT3), c‐kit (CD117) and colony stimulating factor‐1 receptor (CSF1R). Hepatocellular carcinoma (HCC), renal cell carcinoma (RCC) and prostate carcinoma (PC) are known hypervascular/angiogenic tumor types. The role of the vascular process in HCC tumor progression, HIF‐mediated induction of VEGF in many VHL‐deleted RCCs, as well as PDGF expression in PC, highlight the potential of anti angiogenic therapy as an attractive therapeutic strategy. Previous studies have shown potent anti‐tumor and anti‐angiogenic activity against a spectrum of xenografts (HCC, RCC, PC, fibrosarcoma and NSCLC). Thus, the ability of linifanib to inhibit tumor growth in known hypervascular/angiogenic tumors suggests on‐mechanism activity. Linifanib was well tolerated in the clinic and activity was observed in HCC, NSCLC, and renal carcinoma. The ability to be used as combination treatment regimen without excessive impact on tolerability is of great interest to increase clinical efficacy and positively impact patient outcomes. In this study, we sought to evaluate the activity of linifanib in a broad spectrum of xenograft models (breast, colon, HNSCC, liver, NSCLC, SCLC, ovarian and pancreatic), alone or in combination with various cytotoxic therapies (carbotaxol, irinotecan, radiation, 5‐FU/leucovorin, gemcitabine and oxaliplatin). The doses used for ABT‐869 were clinically relevant. For the cytotoxic therapies, doses were determined both by clinical schedules and tolerability as single agents in mice. In all studies, linifanib (25, 12.6, 6.25, p.o., b.i.d.x21) was well tolerated alone or in combination with cytotoxic therapy and demonstrated no exacerbation of cytotoxic agent toxicity as assessed by animal health observations. Significant anti‐tumor efficacy (measured by percent tumor growth inhibition, % TGI) was observed with linifanib monotherapy and the combination with cytotoxic therapies showed significant efficacy over linifanib or cytotoxic therapies alone. Further, lower doses of linifanib in combination with cytotoxic agents achieved similar efficacy as higher doses of linifanib alone. These studies demonstrate the significant activity of linifanib or linifanib in combination with multiple cytotoxic therapies in a spectrum of models representing multiple histological types and provide a rationale for future clinical investigations. Citation Information: Mol Cancer Ther 2009;8(12 Suppl):B14.


Journal of Medicinal Chemistry | 2007

Discovery of N-(4-(3-amino-1H-indazol-4-yl)phenyl)-N'-(2-fluoro-5-methylphenyl)urea (ABT-869), a 3-aminoindazole-based orally active multitargeted receptor tyrosine kinase inhibitor.

Yujia Dai; Kresna Hartandi; Zhiqin Ji; Asma A. Ahmed; Daniel H. Albert; Joy Bauch; Jennifer J. Bouska; Peter F. Bousquet; George A. Cunha; Keith B. Glaser; Christopher M. Harris; Dean Hickman; Jun Guo; Junling Li; Patrick A. Marcotte; Kennan C. Marsh; Maria D. Moskey; Ruth L. Martin; Amanda M. Olson; Donald J. Osterling; Lori J. Pease; Niru B. Soni; Kent D. Stewart; Vincent S. Stoll; Paul Tapang; David R. Reuter; Steven K. Davidsen; Michael R. Michaelides

Collaboration


Dive into the Yujia Dai's collaboration.

Top Co-Authors

Avatar

Steven K. Davidsen

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Keith B. Glaser

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Daniel H. Albert

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar

Patrick A. Marcotte

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Junling Li

University of California

View shared research outputs
Top Co-Authors

Avatar

Paul Tapang

University of California

View shared research outputs
Top Co-Authors

Avatar

Ru-Qi Wei

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge