Network


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

Hotspot


Dive into the research topics where Karen Williams is active.

Publication


Featured researches published by Karen Williams.


Bioorganic & Medicinal Chemistry Letters | 2008

1H-Pyrazolo[3,4-g]hexahydro-isoquinolines as selective glucocorticoid receptor antagonists with high functional activity.

Robin D. Clark; Nicholas C. Ray; Karen Williams; Paul Blaney; Stuart Ward; Peter Crackett; Christopher Hurley; Hazel Joan Dyke; David E. Clark; Peter Lockey; Rene Devos; Melanie Wong; Soraya S. Porres; Colin P. Bright; Robert E. Jenkins; Joseph K. Belanoff

Addition of the 4-fluorophenylpyrazole group to the previously described 2-azadecalin glucocorticoid receptor (GR) antagonist 1 resulted in significantly enhanced functional activity. SAR of the bridgehead substituent indicated that whereas groups as small as methyl afforded high GR binding, GR functional activity was enhanced by larger groups such as benzyl, substituted ethers, and aminoalkyl derivatives. GR antagonists with binding and functional activity comparable to mifepristone were discovered (e.g., 52: GR binding K(i) 0.7 nM; GR reporter gene functional K(i) 0.6 nM) and found to be highly selective over other steroid receptors. Analogues 43 and 45 had >50% oral bioavailability in the dog.


Molecular Cancer Therapeutics | 2013

Bcl-2/Bcl-xL Inhibition Increases the Efficacy of MEK Inhibition Alone and in Combination with PI3 Kinase Inhibition in Lung and Pancreatic Tumor Models

Nguyen Tan; Maureen Wong; Michelle Nannini; Rebecca Hong; Leslie Lee; Stephen Price; Karen Williams; Pierre Pascal Savy; Peng Yue; Deepak Sampath; Jeffrey Settleman; Wayne J. Fairbrother; Lisa D. Belmont

Although mitogen-activated protein (MAP)–extracellular signal-regulated kinase (ERK) kinase (MEK) inhibition is predicted to cause cell death by stabilization of the proapoptotic BH3-only protein BIM, the induction of apoptosis is often modest. To determine if addition of a Bcl-2 family inhibitor could increase the efficacy of a MEK inhibitor, we evaluated a panel of 53 non–small cell lung cancer and pancreatic cancer cell lines with the combination of navitoclax (ABT-263), a Bcl-2/Bcl-xL (BCL2/BCL2L1) antagonist, and a novel MAP kinase (MEK) inhibitor, G-963. The combination is synergistic in the majority of lines, with an enrichment of cell lines harboring KRAS mutations in the high synergy group. Cells exposed to G-963 arrest in G1 and a small fraction undergo apoptosis. The addition of navitoclax to G-963 does not alter the kinetics of cell-cycle arrest, but greatly increases the percentage of cells that undergo apoptosis. The G-963/navitoclax combination was more effective than either single agent in the KRAS mutant H2122 xenograft model; BIM stabilization and PARP cleavage were observed in tumors, consistent with the mechanism of action observed in cell culture. Addition of the phosphatidylinositol 3-kinase (PI3K, PIK3CA) inhibitor GDC-0941 to this treatment combination increases cell killing compared with double- or single-agent treatment. Taken together, these data suggest the efficacy of agents that target the MAPK and PI3K pathways can be improved by combination with a Bcl-2 family inhibitor. Mol Cancer Ther; 12(6); 853–64. ©2013 AACR.


Journal of Immunology | 2013

A Restricted Role for TYK2 Catalytic Activity in Human Cytokine Responses Revealed by Novel TYK2-Selective Inhibitors

Sue J. Sohn; Kathy Barrett; Anne van Abbema; Christine Chang; Pawan Bir Kohli; Hidenobu Kanda; Janice Smith; Yingjie Lai; Aihe Zhou; Birong Zhang; Wenqian Yang; Karen Williams; Calum Macleod; Christopher Hurley; Janusz Jozef Kulagowski; Nicholas Lewin-Koh; Hart S. Dengler; Adam R. Johnson; Nico Ghilardi; Mark Zak; Jun Liang; Wade S. Blair; Steven Magnuson; Lawren C. Wu

TYK2 is a JAK family protein tyrosine kinase activated in response to multiple cytokines, including type I IFNs, IL-6, IL-10, IL-12, and IL-23. Extensive studies of mice that lack TYK2 expression indicate that the IFN-α, IL-12, and IL-23 pathways, but not the IL-6 or IL-10 pathways, are compromised. In contrast, there have been few studies of the role of TYK2 in primary human cells. A genetic mutation at the tyk2 locus that results in a lack of TYK2 protein in a single human patient has been linked to defects in the IFN-α, IL-6, IL-10, IL-12, and IL-23 pathways, suggesting a broad role for TYK2 protein in human cytokine responses. In this article, we have used a panel of novel potent TYK2 small-molecule inhibitors with varying degrees of selectivity against other JAK kinases to address the requirement for TYK2 catalytic activity in cytokine pathways in primary human cells. Our results indicate that the biological processes that require TYK2 catalytic function in humans are restricted to the IL-12 and IL-23 pathways, and suggest that inhibition of TYK2 catalytic activity may be an efficacious approach for the treatment of select autoimmune diseases without broad immunosuppression.


Molecular Cancer Therapeutics | 2013

Combination Drug Scheduling Defines a “Window of Opportunity” for Chemopotentiation of Gemcitabine by an Orally Bioavailable, Selective ChK1 Inhibitor, GNE-900

Elizabeth Blackwood; Jennifer Epler; Ivana Yen; Michael Flagella; Thomas O'Brien; Marie Evangelista; Stephen Schmidt; Yang Xiao; Jonathan Choi; Kaska Kowanetz; Judi Ramiscal; Kenton Wong; Diana Jakubiak; Sharon Yee; Gary Cain; Lewis J. Gazzard; Karen Williams; Jason S. Halladay; Peter K. Jackson; Shiva Malek

Checkpoint kinase 1 (ChK1) is a serine/threonine kinase that functions as a central mediator of the intra-S and G2–M cell-cycle checkpoints. Following DNA damage or replication stress, ChK1-mediated phosphorylation of downstream effectors delays cell-cycle progression so that the damaged genome can be repaired. As a therapeutic strategy, inhibition of ChK1 should potentiate the antitumor effect of chemotherapeutic agents by inactivating the postreplication checkpoint, causing premature entry into mitosis with damaged DNA resulting in mitotic catastrophe. Here, we describe the characterization of GNE-900, an ATP-competitive, selective, and orally bioavailable ChK1 inhibitor. In combination with chemotherapeutic agents, GNE-900 sustains ATR/ATM signaling, enhances DNA damage, and induces apoptotic cell death. The kinetics of checkpoint abrogation seems to be more rapid in p53-mutant cells, resulting in premature mitotic entry and/or accelerated cell death. Importantly, we show that GNE-900 has little single-agent activity in the absence of chemotherapy and does not grossly potentiate the cytotoxicity of gemcitabine in normal bone marrow cells. In vivo scheduling studies show that optimal administration of the ChK1 inhibitor requires a defined lag between gemcitabine and GNE-900 administration. On the refined combination treatment schedule, gemcitabines antitumor activity against chemotolerant xenografts is significantly enhanced and dose-dependent exacerbation of DNA damage correlates with extent of tumor growth inhibition. In summary, we show that in vivo potentiation of gemcitabine activity is mechanism based, with optimal efficacy observed when S-phase arrest and release is followed by checkpoint abrogation with a ChK1 inhibitor. Mol Cancer Ther; 12(10); 1968–80. ©2013 AACR.


Bioorganic & Medicinal Chemistry Letters | 2014

Discovery of the 1,7-diazacarbazole class of inhibitors of checkpoint kinase 1.

Lewis J. Gazzard; Brent A. Appleton; Kerry Chapman; Huifen Chen; Kevin Clark; Joy Drobnick; Simon Goodacre; Jason S. Halladay; Joseph P. Lyssikatos; Stephen Schmidt; Steve Sideris; Christian Wiesmann; Karen Williams; Ping Wu; Ivana Yen; Shiva Malek

Checkpoint kinase 1 (ChK1) is activated in response to DNA damage, acting to temporarily block cell cycle progression and allow for DNA repair. It is envisaged that inhibition of ChK1 will sensitize tumor cells to treatment with DNA-damaging therapies, and may enhance the therapeutic window. High throughput screening identified carboxylate-containing diarylpyrazines as a prominent hit series, but with limited biochemical potency and no cellular activity. Through a series of SAR investigations and X-ray crystallographic analysis the critical role of polar contacts with conserved waters in the kinase back pocket was established. Structure-based design, guided by in silico modeling, transformed the series to better satisfy these contacts and the novel 1,7-diazacarbazole class of inhibitors was discovered. Here we present the genesis of this novel series and the identification of GNE-783, a potent, selective and orally bioavailable inhibitor of ChK1.


ACS Medicinal Chemistry Letters | 2013

Azepines and piperidines with dual norepinephrine dopamine uptake inhibition and antidepressant activity.

Dean G. Brown; Peter R. Bernstein; Ye Wu; Rebecca Urbanek; Christopher Becker; Scott Throner; Bruce T. Dembofsky; Gary Steelman; Lois Ann Lazor; Clay W Scott; Michael W. Wood; Steven Wesolowski; David A. Nugiel; Stephanie Koch; Jian Yu; Donald E. Pivonka; Shuang Li; Carol Thompson; Anna Zacco; Charles S. Elmore; Patricia Schroeder; Jianwei Liu; Christopher Hurley; Stuart Ward; Hazel J. Hunt; Karen Williams; Joseph McLaughlin; Valerie Hoesch; Simon Sydserff; Donna L. Maier

Herein, we describe the discovery of inhibitors of norepinephrine (NET) and dopamine (DAT) transporters with reduced activity relative to serotonin transporters (SERT). Two compounds, 8b and 21a, along with nomifensine were tested in a rodent receptor occupancy study and demonstrated dose-dependent displacement of radiolabeled NET and DAT ligands. These compounds were efficacious in a rat forced swim assay (model of depression) and also had activity in rat spontaneous locomotion assay.


Journal of Medicinal Chemistry | 2015

Mitigation of Acetylcholine Esterase Activity in the 1,7-Diazacarbazole Series of Inhibitors of Checkpoint Kinase 1

Lewis J. Gazzard; Karen Williams; Huifen Chen; Lorraine Axford; Elizabeth Blackwood; Brenda Burton; Kerry L. Chapman; Peter Crackett; Joy Drobnick; Charles Ellwood; Jennifer Epler; Michael Flagella; Emanuela Gancia; Matthew Gill; Simon Charles Goodacre; Jason S. Halladay; Joanne Frances Mary Hewitt; Hazel J. Hunt; Samuel Kintz; Joseph P. Lyssikatos; Calum Macleod; Sarah Major; Guillaume Médard; Raman Narukulla; Judi Ramiscal; Stephen Schmidt; Eileen Seward; Christian Wiesmann; Ping Wu; Sharon Yee

Checkpoint kinase 1 (ChK1) plays a key role in the DNA damage response, facilitating cell-cycle arrest to provide sufficient time for lesion repair. This leads to the hypothesis that inhibition of ChK1 might enhance the effectiveness of DNA-damaging therapies in the treatment of cancer. Lead compound 1 (GNE-783), the prototype of the 1,7-diazacarbazole class of ChK1 inhibitors, was found to be a highly potent inhibitor of acetylcholine esterase (AChE) and unsuitable for development. A campaign of analogue synthesis established SAR delineating ChK1 and AChE activities and allowing identification of new leads with improved profiles. In silico docking using a model of AChE permitted rationalization of the observed SAR. Compounds 19 (GNE-900) and 30 (GNE-145) were identified as selective, orally bioavailable ChK1 inhibitors offering excellent in vitro potency with significantly reduced AChE activity. In combination with gemcitabine, these compounds demonstrate an in vivo pharmacodynamic effect and are efficacious in a mouse p53 mutant xenograft model.


Bioorganic & Medicinal Chemistry Letters | 2017

Identification of an imidazopyridine scaffold to generate potent and selective TYK2 inhibitors that demonstrate activity in an in vivo psoriasis model.

Jun Liang; Anne van Abbema; Mercedesz Balazs; Kathy Barrett; Leo Berezhkovsky; Wade S. Blair; Christine Chang; Donnie Delarosa; Jason DeVoss; Jim Driscoll; Charles Eigenbrot; Simon Charles Goodacre; Nico Ghilardi; Calum Macleod; Adam R. Johnson; Pawan Bir Kohli; Yingjie Lai; Zhonghua Lin; Priscilla Mantik; Kapil Menghrajani; Hieu Nguyen; Ivan Peng; Amy Sambrone; Steven Shia; Jan Smith; Sue Sohn; Vickie Tsui; Mark Ultsch; Karen Williams; Lawren C. Wu

Herein we report identification of an imidazopyridine class of potent and selective TYK2 inhibitors, exemplified by prototype 6, through constraint of the rotatable amide bond connecting the pyridine and aryl rings of compound 1. Further optimization led to generation of compound 30 that potently inhibits the TYK2 enzyme and the IL-23 pathway in cells, exhibits selectivity against cellular JAK2 activity, and has good pharmacokinetic properties. In mice, compound 30 demonstrated dose-dependent reduction of IL-17 production in a PK/PD model as well as in an imiquimod-induced psoriasis model. In this efficacy model, the IL-17 decrease was accompanied by a reduction of ear thickness indicating the potential of TYK2 inhibition as a therapeutic approach for psoriasis patients.


Bioorganic & Medicinal Chemistry Letters | 2007

Discovery and optimization of novel, non-steroidal glucocorticoid receptor modulators

Nicholas C. Ray; Robin D. Clark; David E. Clark; Karen Williams; H.G. Hickin; Peter Crackett; Hazel Joan Dyke; Peter Lockey; Melanie Wong; Rene Devos; Anne White; Joseph K. Belanoff


Archive | 2005

Fused ring azadecalin glucocorticoid receptor modulators

Robin D. Clark; Nicholas Charles Ray; Paul Blaney; Christopher Hurley; Karen Williams

Collaboration


Dive into the Karen Williams'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
Researchain Logo
Decentralizing Knowledge