Network


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

Hotspot


Dive into the research topics where Lance H. Ridgers is active.

Publication


Featured researches published by Lance H. Ridgers.


ACS Medicinal Chemistry Letters | 2010

Discovery of GSK2126458, a Highly Potent Inhibitor of PI3K and the Mammalian Target of Rapamycin.

Steven David Knight; Nicholas D. Adams; Joelle L. Burgess; Amita M. Chaudhari; Michael G. Darcy; Carla A. Donatelli; Juan I. Luengo; Ken A. Newlander; Cynthia A. Parrish; Lance H. Ridgers; Martha A. Sarpong; Stanley J. Schmidt; Glenn S. Van Aller; Jeffrey D. Carson; Melody Diamond; Patricia A. Elkins; Christine M. Gardiner; Eric Garver; Seth Gilbert; Richard R. Gontarek; Jeffrey R. Jackson; Kevin L. Kershner; Lusong Luo; Kaushik Raha; Christian S. Sherk; Chiu-Mei Sung; David Sutton; Peter J. Tummino; Ronald Wegrzyn; Kurt R. Auger

Phosphoinositide 3-kinase α (PI3Kα) is a critical regulator of cell growth and transformation, and its signaling pathway is the most commonly mutated pathway in human cancers. The mammalian target of rapamycin (mTOR), a class IV PI3K protein kinase, is also a central regulator of cell growth, and mTOR inhibitors are believed to augment the antiproliferative efficacy of PI3K/AKT pathway inhibition. 2,4-Difluoro-N-{2-(methyloxy)-5-[4-(4-pyridazinyl)-6-quinolinyl]-3-pyridinyl}benzenesulfonamide (GSK2126458, 1) has been identified as a highly potent, orally bioavailable inhibitor of PI3Kα and mTOR with in vivo activity in both pharmacodynamic and tumor growth efficacy models. Compound 1 is currently being evaluated in human clinical trials for the treatment of cancer.


British Journal of Pharmacology | 2005

Nonpeptidic urotensin-II receptor antagonists I: in vitro pharmacological characterization of SB-706375

Stephen A. Douglas; David J. Behm; Nambi Aiyar; Diane Naselsky; Jyoti Disa; David P. Brooks; Eliot H. Ohlstein; John G Gleason; Henry M. Sarau; James J. Foley; Peter T. Buckley; Dulcie B. Schmidt; William E. Wixted; Katherine L. Widdowson; Graham J. Riley; Jian Jin; Timothy F. Gallagher; Stanley J. Schmidt; Lance H. Ridgers; Lisa T. Christmann; Richard M. Keenan; Steven D. Knight; Dashyant Dhanak

1 SB‐706375 potently inhibited [125I]hU‐II binding to both mammalian recombinant and ‘native’ UT receptors (Ki 4.7±1.5 to 20.7±3.6 nM at rodent, feline and primate recombinant UT receptors and Ki 5.4±0.4 nM at the endogenous UT receptor in SJRH30 cells). 2 Prior exposure to SB‐706375 (1 μM, 30 min) did not alter [125I]hU‐II binding affinity or density in recombinant cells (KD 3.1±0.4 vs 5.8±0.9 nM and Bmax 3.1±1.0 vs 2.8±0.8 pmol mg−1) consistent with a reversible mode of action. 3 The novel, nonpeptidic radioligand [3H]SB‐657510, a close analogue of SB‐706375, bound to the monkey UT receptor (KD 2.6±0.4 nM, Bmax 0.86±0.12 pmol mg−1) in a manner that was inhibited by both U‐II isopeptides and SB‐706375 (Ki 4.6±1.4 to 17.6±5.4 nM) consistent with the sulphonamides and native U‐II ligands sharing a common UT receptor binding domain. 4 SB‐706375 was a potent, competitive hU‐II antagonist across species with pKb 7.29–8.00 in HEK293‐UT receptor cells (inhibition of [Ca2+]i‐mobilization) and pKb 7.47 in rat isolated aorta (inhibition of contraction). SB‐706375 also reversed tone established in the rat aorta by prior exposure to hU‐II (Kapp∼20 nM). 5 SB‐706375 was a selective U‐II antagonist with 100‐fold selectivity for the human UT receptor compared to 86 distinct receptors, ion channels, enzymes, transporters and nuclear hormones (Ki/IC50>1 μM). Accordingly, the contractile responses induced in isolated aortae by KCl, phenylephrine, angiotensin II and endothelin‐1 were unaltered by SB‐706375 (1 μM). 6 In summary, SB‐706375 is a high‐affinity, surmountable, reversible and selective nonpeptide UT receptor antagonist with cross‐species activity that will assist in delineating the pathophysiological actions of U‐II in mammals.


Bioorganic & Medicinal Chemistry Letters | 2001

Discovery of potent and selective phenylalanine derived CCR3 receptor antagonists. Part 2.

Dashyant Dhanak; Lisa T. Christmann; Michael G. Darcy; Richard M. Keenan; Steven David Knight; Judithann M. Lee; Lance H. Ridgers; Henry M. Sarau; Dinubhai H Shah; John R. White; Lily Zhang

Highly potent CCR3 antagonists have been developed from a previously reported series of phenylalanine ester-based leads. Solution-phase, parallel synthesis optimization was utilized to identify highly potent, functional CCR3 antagonists.


ACS Medicinal Chemistry Letters | 2010

Discovery of the First Potent and Selective Inhibitor of Centromere-Associated Protein E: GSK923295.

Xiangping Qian; Andrew Mcdonald; Han-Jie Zhou; Nicholas D. Adams; Cynthia A. Parrish; Kevin J. Duffy; Duke M. Fitch; Rosanna Tedesco; Luke W. Ashcraft; Bing Yao; Hong Jiang; Jennifer Kuo Chen Huang; Melchor V. Marin; Carrie E. Aroyan; Jianchao Wang; Seyed Ahmed; Joelle L. Burgess; Amita M. Chaudhari; Carla A. Donatelli; Michael G. Darcy; Lance H. Ridgers; Ken A. Newlander; Stanley J. Schmidt; Deping Chai; Mariela Colón; Michael N. Zimmerman; Latesh Lad; Roman Sakowicz; Stephen Schauer; Lisa Belmont

Inhibition of mitotic kinesins represents a novel approach for the discovery of a new generation of anti-mitotic cancer chemotherapeutics. We report here the discovery of the first potent and selective inhibitor of centromere-associated protein E (CENP-E) 3-chloro-N-{(1S)-2-[(N,N-dimethylglycyl)amino]-1-[(4-{8-[(1S)-1-hydroxyethyl]imidazo[1,2-a]pyridin-2-yl}phenyl)methyl]ethyl}-4-[(1-methylethyl)oxy]benzamide (GSK923295; 1), starting from a high-throughput screening hit, 3-chloro-4-isopropoxybenzoic acid 2. Compound 1 has demonstrated broad antitumor activity in vivo and is currently in human clinical trials.


Bioorganic & Medicinal Chemistry Letters | 1998

Benzothiopyran-4-one based reversible inhibitors of the human cytomegalovirus (HCMV) protease

Dashyant Dhanak; Richard M. Keenan; George Burton; Arun C. Kaura; Michael G. Darcy; Dinubhai H Shah; Lance H. Ridgers; Ann L. Breen; Paddy Lavery; David G. Tew; Andrew West

A novel class of CMV protease inhibitors based on a benzothiopyran-S,S-dioxide nucleus has been discovered. Enzyme kinetic data supports a reversible mode of inhibition for a representative member of this class, 2-(3-pyridyl-N-oxide)benzothiopyran-4-one-S,S-dioxide, 1. Experiments in the presence and absence of the disulfide reducing agent DTT suggest that the inhibition by 1 is not due to oxidative inactivation of the enzyme. Also presented are results of some SAR studies of the benzothiopyranone ring system.


Bioorganic & Medicinal Chemistry Letters | 1999

Design and synthesis of diaminopyrrolidinone inhibitors of human osteoclast cathepsin K

Kevin J. Duffy; Lance H. Ridgers; Renee L. DesJarlais; Thaddeus A. Tomaszek; Mary J. Bossard; Scott K. Thompson; Richard M. Keenan; Daniel F. Veber

The structure-based design and synthesis of lactam-constrained azapeptide inhibitors of human cathepsin K are described. Enhanced stability to proteolytic cleavage over acyclic analogues is discussed.


Molecular Cancer Therapeutics | 2009

Abstract C62: Identification of GSK2126458, a highly potent inhibitor of phosphoinositide 3‐kinase (PI3K) and the mammalian target of rapamycin (mTOR)

Steven D. Knight; Nicholas D. Adams; Joelle L. Burgess; Amita M. Chaudhari; Michael G. Darcy; Carla A. Donatelli; Ken Newlander; Cynthia A. Parrish; Lance H. Ridgers; Martha A. Sarpong; Stanley J. Schmidt; Glenn S. Van Aller; Jeffrey D. Carson; Patricia A. Elkins; Melody Diamond; Christine M. Gardiner; Eric Garver; Lusong Luo; Kaushik Raha; Chiu-Mei Sung; Peter J. Tummino; Kurt R. Auger; Dashyant Dhanak

Phosphoinositide 3‐kinase (PI3K) is a critical regulator of cell growth and transformation and its signaling pathway is one of the most commonly mutated pathways in human cancer. The mammalian target of rapamycin (mTOR), a class IV PI3K protein kinase, is also a central regulator of cell growth, and mTOR inhibitors are believed to augment the antiproliferative efficacy of the PI3K/AKT pathway. GSK1059615, our first PI3K clinical compound, progressed to a dose escalation study in patients with refractory malignancies. Following the discovery of GSK1059615, we sought to identify a second inhibitor with improved potency, selectivity, and pharmacokinetics. Key to our approach to achieving the desired levels of PI3K activity was to pursue structure‐based design utilizing crystallography of the more amenable PI3K as a surrogate protein. Following a chemistry lead optimization effort, the pyridylsulfonamide GSK2126458 was identified as a highly potent, orally bioavailable, pan‐PI3K and mTOR inhibitor (PI3K app Ki = 19 pM; mTORC1 app Ki = 180 pM; mTORC2 app Ki = 300 pM). Consistent with potent PI3K and mTORC2 enzyme inhibition, GSK2126458 decreased cellular levels of phosphorylated AKT (BT474 pAKT IC50 = 180 pM) and inhibited cell proliferation in a large panel of cancer cell lines (e.g. BT474 growth IC50 = 2 nM). GSK2126458 showed good exposure in four pre‐clinical animal species and exhibited in vivo activity in both pharmacodynamic and tumor growth efficacy models. GSK2126458 is being evaluated currently in human clinical trials for the treatment of cancer. The discovery, design, and optimization of GSK2126458 and related analogs will be presented. Citation Information: Mol Cancer Ther 2009;8(12 Suppl):C62.


Journal of Medicinal Chemistry | 2007

Novel ATP-competitive kinesin spindle protein inhibitors

Cynthia A. Parrish; Nicholas D. Adams; Kurt R. Auger; Joelle L. Burgess; Jeffrey D. Carson; Amita M. Chaudhari; Robert A. Copeland; Melody Diamond; Carla A. Donatelli; Kevin J. Duffy; Leo F. Faucette; Jeffrey T. Finer; William F. Huffman; Erin D. Hugger; Jeffrey R. Jackson; Steven David Knight; Lusong Luo; Michael L. Moore; Ken A. Newlander; Lance H. Ridgers; Roman Sakowicz; Antony N. Shaw; Chiu-Mei M. Sung; David Sutton; Kenneth W. Wood; Shu-Yun Zhang; Michael N. Zimmerman; Dashyant Dhanak


Archive | 2008

Quinoline derivatives as p13 kinase inhibitors

Nicholas D. Adams; Joelle L. Burgess; Michael G. Darcy; Carla A. Donatelli; Steven D. Knight; Kenneth A. Newlander; Lance H. Ridgers; Martha A. Sarpong; Stanley J. Schmidt


Archive | 2008

Quinoline derivatives as pi3 kinase inhibitors

Nicholas D. Adams; Joelle L. Burgess; Michael Gerrard Darcy; Carla A. Donatelli; Steven David Knight; Kenneth A. Newlander; Lance H. Ridgers; Martha A. Sarpong; Stanley J. Schmidt

Collaboration


Dive into the Lance H. Ridgers's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge