Network


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

Hotspot


Dive into the research topics where Suining Lee is active.

Publication


Featured researches published by Suining Lee.


Journal of Medicinal Chemistry | 2011

Aurora Kinase Inhibitors Based on the Imidazo[1,2-a]pyrazine Core: Fluorine and Deuterium Incorporation Improve Oral Absorption and Exposure

Angela Kerekes; Sara Esposite; Ronald J. Doll; Jayaram R. Tagat; Tao Yu; Yushi Xiao; Yonglian Zhang; Dan Prelusky; Seema Tevar; Kimberly Gray; Gaby A. Terracina; Suining Lee; Jennifer Jones; Ming Liu; Andrea Basso; Elizabeth B. Smith

Aurora kinases are cell cycle regulated serine/threonine kinases that have been linked to cancer. Compound 1 was identified as a potent Aurora inhibitor but lacked oral bioavailability. Optimization of 1 led to the discovery of a series of fluoroamine and deuterated analogues, exemplified by compound 25, with an improved pharmacokinetic profile. We found that blocking oxidative metabolism at the benzylic position and decreasing the basicity of the amine are important to obtaining compounds with good biological profiles and oral bioavailability.


ACS Medicinal Chemistry Letters | 2010

Discovery of a Potent, Injectable Inhibitor of Aurora Kinases Based on the Imidazo-[1,2-a]-Pyrazine Core

Tao Yu; Jayaram R. Tagat; Angela Kerekes; Ronald J. Doll; Yonglian Zhang; Yushi Xiao; Sara Esposite; David B. Belanger; Patrick J. Curran; Amit K. Mandal; M. Arshad Siddiqui; Neng-Yang Shih; Andrea D. Basso; Ming Liu; Kimberly Gray; Seema Tevar; Jennifer Jones; Suining Lee; Lianzhu Liang; Samad Ponery; Elizabeth B. Smith; Alan Hruza; Johannes Voigt; Lata Ramanathan; Winifred W. Prosise; Mengwei Hu

The imidazo-[1,2-a]-pyrazine (1) is a dual inhibitor of Aurora kinases A and B with modest cell potency (IC50 = 250 nM) and low solubility (5 μM). Lead optimization guided by the binding mode led to the acyclic amino alcohol 12k (SCH 1473759), which is a picomolar inhibitor of Aurora kinases (TdF K d Aur A = 0.02 nM and Aur B = 0.03 nM) with improved cell potency (phos-HH3 inhibition IC50 = 25 nM) and intrinsic aqueous solubility (11.4 mM). It also demonstrated efficacy and target engagement in human tumor xenograft mouse models.


Bioorganic & Medicinal Chemistry Letters | 2010

Discovery of orally bioavailable imidazo[1,2-a]pyrazine-based Aurora kinase inhibitors.

David B. Belanger; Michael Williams; Patrick J. Curran; Amit K. Mandal; Zhaoyang Meng; Matthew P. Rainka; Tao Yu; Neng-Yang Shih; M. Arshad Siddiqui; Ming Liu; Seema Tevar; Suining Lee; Lianzhu Liang; Kimberly Gray; Bohdan Yaremko; Jennifer Jones; Elizabeth B. Smith; Dan Prelusky; Andrea D. Basso

We report a series of potent imidazo[1,2-a]pyrazine-based Aurora kinase inhibitors. Optimization of the solvent accessible 8-position led to improvements in both oral bioavailability and off-target kinase inhibition. Compound 25 demonstrates anti-tumor activity in an A2780 ovarian tumor xenograft model.


Molecular Cancer Therapeutics | 2010

SCH 2047069, a Novel Oral Kinesin Spindle Protein Inhibitor, Shows Single-Agent Antitumor Activity and Enhances the Efficacy of Chemotherapeutics

Andrea D. Basso; Ming Liu; Chaoyang Dai; Kimberly Gray; Lissette Nale; Seema Tevar; Suining Lee; Lianzhu Liang; Abdul Ponery; Bohdan Yaremko; Elizabeth M. Smith; Huadong Tang; Payal R. Sheth; M. Arshad Siddiqui; Daniel J. Hicklin; Paul Kirschmeier

Kinesin spindle protein (KSP) is a mitotic kinesin required for the formation of the bipolar mitotic spindle, and inhibition of this motor protein results in mitotic arrest and cell death. KSP inhibitors show preclinical antitumor activity and are currently undergoing testing in clinical trials. These agents have been dosed intravenously using various dosing schedules. We sought to identify a KSP inhibitor that could be delivered orally and thus provide convenience of dosing as well as the ability to achieve more continuous exposure via the use of dose-dense administration. We discovered SCH 2047069, a potent KSP inhibitor with oral bioavailability across species and the ability to cross the blood-brain barrier. The compound induces mitotic arrest characterized by a monaster spindle and is associated with an increase in histone H3 and mitotic protein monoclonal 2 phosphorylation both in vitro and in vivo. SCH 2047069 showed antitumor activity in a variety of preclinical models as a single agent and in combination with paclitaxel, gemcitabine, or vincristine. Mol Cancer Ther; 9(11); 2993–3002. ©2010 AACR.


Cancer Research | 2010

Abstract 1648: SCH 1473759, a novel aurora inhibitor, demonstrates enhanced antitumor activity in combination with taxanes and KSP inhibitors

Andrea D. Basso; Kimberly Gray; Seema Tevar; Samad Ponery; Suining Lee; Ming Liu; Elizabeth M. Smith; Tao Yu; Jay Tagat; Ronald J. Doll; David B. Belanger; Arshad Siddiqui; Yonglain Zhang; Yushi Xiao; Sara Esposite; Fredrick Monsma; Dan Hicklin; Paul Kirschmeier

Aurora kinases are required for orderly progression of cells through mitosis. Inhibition of these kinases by siRNA or a small molecule inhibitors results in aberrant endoreduplication and cell death. SCH 1473759 is a novel Aurora inhibitor with potent mechanism based cell activity. The compound is active against a large panel of tumor cell lines from different tissue origin and genetic backgrounds. We found that asynchronous cells require 24 hour exposure to SCH 1473759 to induce maximal endoreduplication and cell kill. However, following a taxane or KSP inhibitor mitotic arrest, less than 4-hour exposure was sufficient to induce endoreduplication. This finding correlated with the ability of SCH 1473759 to accelerate exit from mitosis in response to taxane and KSP induced arrest, but not that of a nocodazole arrest. SCH 1473759 demonstrated single agent biomarker and anti-tumor activity in A2780 ovarian xenograft models. Further, efficacy was enhanced in combination with taxotere and found to be most efficacious when SCH 1473759 was dosed 12-hours post taxotere. These findings could have clinical implications for the development of Aurora inhibitors. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 1648.


Cancer Research | 1998

Antitumor activity of SCH 66336, an orally bioavailable tricyclic inhibitor of farnesyl protein transferase, in human tumor xenograft models and wap-ras transgenic mice.

Ming Liu; Matthew S. Bryant; Jianping Chen; Suining Lee; Bohdan Yaremko; Phil Lipari; Michael Malkowski; Eric Ferrari; Loretta L. Nielsen; Nicholas Prioli; Janet Dell; Dineshwar Sinha; Jameel Syed; Walter A. Korfmacher; Amin A. Nomeir; C-C. Lin; Lynn Wang; Arthur G. Taveras; Ronald J. Doll; F. George Njoroge; Alan K. Mallams; Stacy W. Remiszewski; Joseph J. Catino; Viyyoor M. Girijavallabhan; Paul Kirschmeier; W. Robert Bishop


Journal of Medicinal Chemistry | 1997

Structure-activity relationship of 3-substituted N-(pyridinylacetyl)-4-(8-chloro-5,6-dihydro-11H-benzo[5,6]cyclohepta[1,2-b]pyridin-11-ylidene)-piperidine inhibitors of farnesyl-protein transferase : Design and synthesis of in vivo active antitumor compounds

Njoroge Fg; Bancha Vibulbhan; Rane Df; Bishop Wr; Joanne M. Petrin; Robert Patton; Mathew S. Bryant; Kwang-jong Chen; Amin A. Nomeir; Chin-Chung Lin; Ming Liu; King I; Jianping Chen; Suining Lee; Yaremko B; Janet Dell; Philip Lipari; Michael Malkowski; Zujun Li; Joseph J. Catino; Ronald J. Doll; Girijavallabhan; Ashit K. Ganguly


Journal of Medicinal Chemistry | 1998

Inhibitors of Farnesyl Protein Transferase. 4-Amido, 4-Carbamoyl, and 4-Carboxamido Derivatives of 1-(8-Chloro-6,11-dihydro-5H-benzo[5,6]- cyclohepta[1,2-b]pyridin-11-yl)piperazine and 1-(3-Bromo-8-chloro-6,11- dihydro-5H-benzo[5,6]cyclohepta[1,2-b]pyridin-11-yl)piperazine1

Alan K. Mallams; Randall R. Rossman; Ronald J. Doll; Viyyoor M. Girijavallabhan; Ashit K. Ganguly; Joanne M. Petrin; Lynn Wang; Robert Patton; W. Robert Bishop; Donna Carr; Paul Kirschmeier; Joseph J. Catino; Matthew Bryant; Kwang-jong Chen; Walter A. Korfmacher; Cymbelene Nardo; Shiyong Wang; Amin A. Nomeir; Chin-Chung Lin; Zujun Li; Jianping Chen; Suining Lee; Janet Dell; Philip Lipari; Michael Malkowski; Bodan Yaremko; and Ivan King; Ming Liu


Cancer Chemotherapy and Pharmacology | 2011

SCH 1473759, a novel Aurora inhibitor, demonstrates enhanced anti-tumor activity in combination with taxanes and KSP inhibitors

Andrea D. Basso; Ming Liu; Kimberly Gray; Seema Tevar; Suining Lee; Lianzhu Liang; Abdul Ponery; Elizabeth B. Smith; Frederick J. Monsma; Tao Yu; Yonglian Zhang; Angela Kerekes; Sara Esposite; Yushi Xiao; Jayaram R. Tagat; Daniel J. Hicklin; Paul Kirschmeier


ACS Medicinal Chemistry Letters | 2010

Discovery of a Potent and Injectable Inhibitor of Aurora Kinases A and B based on the Imidazo-[1, 2-a]-Pyrazine Core

Tao Yu; Jayaram R. Tagat; Angela Kerekes; Ronald J. Doll; Yonglian Zhang; Yushi Xiao; Sara Esposite; David B. Belanger; Patrick J. Curran; Amit K. Mandal; M.A Siddiqui; Neng-Yang Shih; Andrea D. Basso; Ming Liu; Kimberly Gray; Seema Tevar; Jennifer Jones; Suining Lee; Lianzhu Liang; Samad Ponery; Elizabeth B. Smith; Alan Hruza; Johannes Voigt; Lata Ramanathan; Winifred W. Prosise; M. Hu

Collaboration


Dive into the Suining Lee's collaboration.

Researchain Logo
Decentralizing Knowledge