Steven John Woodhead
Astex
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Steven John Woodhead.
Clinical Cancer Research | 2012
Timothy A. Yap; Mike I. Walton; Kyla Grimshaw; Robert te Poele; Paul D. Eve; Melanie Valenti; Alexis de Haven Brandon; Vanessa Martins; Anna Zetterlund; Simon P. Heaton; Kathrin Heinzmann; Paul S. Jones; Ruth Feltell; Matthias Reule; Steven John Woodhead; Thomas G. Davies; John Lyons; Florence I. Raynaud; Suzanne A. Eccles; Paul Workman; Neil Thomas Thompson; Michelle D. Garrett
Purpose: Deregulated phosphatidylinositol 3-kinase pathway signaling through AGC kinases including AKT, p70S6 kinase, PKA, SGK and Rho kinase is a key driver of multiple cancers. The simultaneous inhibition of multiple AGC kinases may increase antitumor activity and minimize clinical resistance compared with a single pathway component. Experimental Design: We investigated the detailed pharmacology and antitumor activity of the novel clinical drug candidate AT13148, an oral ATP-competitive multi-AGC kinase inhibitor. Gene expression microarray studies were undertaken to characterize the molecular mechanisms of action of AT13148. Results: AT13148 caused substantial blockade of AKT, p70S6K, PKA, ROCK, and SGK substrate phosphorylation and induced apoptosis in a concentration and time-dependent manner in cancer cells with clinically relevant genetic defects in vitro and in vivo. Antitumor efficacy in HER2-positive, PIK3CA-mutant BT474 breast, PTEN-deficient PC3 human prostate cancer, and PTEN-deficient MES-SA uterine tumor xenografts was shown. We show for the first time that induction of AKT phosphorylation at serine 473 by AT13148, as reported for other ATP-competitive inhibitors of AKT, is not a therapeutically relevant reactivation step. Gene expression studies showed that AT13148 has a predominant effect on apoptosis genes, whereas the selective AKT inhibitor CCT128930 modulates cell-cycle genes. Induction of upstream regulators including IRS2 and PIK3IP1 as a result of compensatory feedback loops was observed. Conclusions: The clinical candidate AT13148 is a novel oral multi-AGC kinase inhibitor with potent pharmacodynamic and antitumor activity, which shows a distinct mechanism of action from other AKT inhibitors. AT13148 will now be assessed in a first-in-human phase I trial. Clin Cancer Res; 18(14); 3912–23. ©2012 AACR.
Molecular Cancer Therapeutics | 2010
Kyla Grimshaw; Lisa-Jane K. Hunter; Timothy A. Yap; Simon P. Heaton; Mike I. Walton; Steven John Woodhead; Lynsey Fazal; Matthias Reule; Thomas G. Davies; Lisa Seavers; Victoria Lock; John Lyons; Neil Thomas Thompson; Paul Workman; Michelle D. Garrett
The serine/threonine kinase AKT plays a pivotal role in signal transduction events involved in malignant transformation and chemoresistance and is an attractive target for the development of cancer therapeutics. Fragment-based lead discovery, combined with structure-based drug design, has recently identified AT7867 as a novel and potent inhibitor of both AKT and the downstream kinase p70 S6 kinase (p70S6K) and also of protein kinase A. This ATP-competitive small molecule potently inhibits both AKT and p70S6K activity at the cellular level, as measured by inhibition of GSK3β and S6 ribosomal protein phosphorylation, and also causes growth inhibition in a range of human cancer cell lines as a single agent. Induction of apoptosis was detected by multiple methods in tumor cells following AT7867 treatment. Administration of AT7867 (90 mg/kg p.o. or 20 mg/kg i.p.) to athymic mice implanted with the PTEN-deficient U87MG human glioblastoma xenograft model caused inhibition of phosphorylation of downstream substrates of both AKT and p70S6K and induction of apoptosis, confirming the observations made in vitro. These doses of AT7867 also resulted in inhibition of human tumor growth in PTEN-deficient xenograft models. These data suggest that the novel strategy of AKT and p70S6K blockade may have therapeutic value and supports further evaluation of AT7867 as a single-agent anticancer strategy. Mol Cancer Ther; 9(5); 1100–10. ©2010 AACR.
Current Topics in Medicinal Chemistry | 2009
Thomas G. Davies; Steven John Woodhead; Ian Collins
Multiple ligand efficient fragment inhibitors of protein kinase B were identified through a combined in silico compound screen and high-throughput crystallographic analysis of protein-ligand structures. A well-validated apo-PKB-PKA chimeric protein provided a convenient platform for high-throughput crystallography by soaking of inhibitors, and a method for the determination of PKB-ligand structures was developed to support inhibitor development. Pyrazole and azaindole fragment hits with micromolar activity were rapidly progressed to nanomolar inhibitors of PKB with activity in cells using crystallographic analysis of inhibitor binding modes to guide medicinal chemistry. Compounds with selectivity for PKB over PKA and other kinases were identified by this approach, resulting in potent inhibitors with in vivo activity through both oral and systemic administration, and suitable for further development.
Cancer Research | 2014
Patrick Angibaud; Laurence Anne Mevellec; Gordon Saxty; Christophe Adelinet; Rhalid Akkari; Valerio Berdini; Pascal Bonnet; Marine Bourgeois; Xavier Bourdrez; Anne Cleasby; Hélène Colombel; Imre Csoka; Werner Constant Johan Embrechts; Eddy Jean Edgard Freyne; Ronaldus Arnodus Hendrika Joseph Gilissen; Eleonora Jovcheva; Peter King; Jean Fernand Armand Lacrampe; Delphine Yvonne Raymonde Lardeau; Yannick Ligny; Steve Mcclue; Lieven Meerpoel; David R. Newell; Martin Page; Alexandra Papanikos; Elisabeth Pasquier; Isabelle Pilatte; Virginie Poncelet; Olivier Querolle; David C. Rees
Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA Fibroblast growth factors (FGFs) and their receptors (FGFR1 through 4) regulate a variety of key cellular processes, including proliferation, migration, survival, and differentiationa. Aberrant activation of FGF/FGFR is strongly implicated in oncogenic signalling in many tumor types. This has stimulated the development of a number of FGFR inhibitors, with diverse kinase inhibition and pharmacological profiles that are currently being evaluated in clinical studies. We conducted a fragment screening campaign and this resulted in identification of a 6-aminoquinoxalinyl fragment with a binding affinity in the micromolar range. Structure-guided medicinal chemistry led to the identification of a novel quinoxaline-based chemical series with nanomolar affinity for FGFR1, 2, 3, and 4, activity in cells, and selectivity with respect to VEGFR-2. Further optimisation resulted in the generation of JNJ-42756493, a compound with favourable drug-like properties that demonstrated strong anti-tumoral activity in a FGFR2-dependent SNU-16 human gastric carcinoma xenograft model. This report represents the first disclosure of the structure-activity relationships as well as the chemical synthesis pathway of the JNJ-42756493 series and illustrates how a fragment-based drug discovery approach has been efficiently used to discover FGFR1-4 inhibitors with nanomolar affinity. aTurner, N. and Grose, R. Nat. Rev. Cancer, 2010, 10, 116-129. Citation Format: Patrick R. Angibaud, Laurence Mevellec, Gordon Saxty, Christophe Adelinet, Rhalid Akkari, Valerio Berdini, Pascal Bonnet, Marine Bourgeois, Xavier Bourdrez, Anne Cleasby, Helene Colombel, Imre Csoka, Werner Embrechts, Eddy Freyne, Ronaldus Gilissen, Eleonora Jovcheva, Peter King, Jean Lacrampe, Delphine Lardeau, Yannick Ligny, Steve Mcclue, Lieven Meerpoel, David R. Newell, Martin Page, Alexandra Papanikos, Elisabeth Pasquier, Isabelle Pilatte, Virginie Poncelet, Olivier Querolle, David C. Rees, Sharna Rich, Bruno Roux, Elodie Sement, Yvan Simonnet, Matthew Squires, Virginie Tronel, Tinne Verhulst, Jorge Vialard, Marc Willems, Steven J. Woodhead, Berthold Wroblowski, Christopher W. Murray, Timothy Perera. Discovery of JNJ-42756493, a potent fibroblast growth factor receptor (FGFR) inhibitor using a fragment based approach. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 4748. doi:10.1158/1538-7445.AM2014-4748
Cancer Research | 2012
Timothy A. Yap; Michael I. Walton; Kyla M. Grimshaw; Robert te Poele; Paul D. Eve; Melanie Valenti; Alexis de Haven Brandon; Vanessa Martins; Anna Zetterlund; Simon P. Heaton; Kathrin Heinzmann; Paul Jones; Ruth Feltell; Matthias Reule; Steven John Woodhead; Thomas G. Davies; John Lyons; Florence I. Raynaud; Suzanne A. Eccles; Paul Workman; Neil Thompson; Michelle D. Garrett
Proceedings: AACR 103rd Annual Meeting 2012‐‐ Mar 31‐Apr 4, 2012; Chicago, IL The AGC kinase AKT is a key component of the phosphatidylinositol 3-kinase (PI3K) pathway, which is frequently deregulated in cancer, making AKT a target of major therapeutic interest. However, PI3K signaling through both AKT-dependent and AKT-independent mechanisms involving other AGC kinases, such as p70S6K, PKA, SGK and ROCK, is important in a range of cancers. Hence, the pharmacological inhibition of these multiple AGC kinases may increase response rates and minimize clinical resistance compared with targeting AKT alone. The clinical drug candidate AT13148 is a multi-AGC kinase, ATP-competitive inhibitor, identified utilizing high-throughput X-ray crystallography and fragment-based lead discovery techniques. Screening of this oral small molecule against a panel of kinases at 10μM revealed >80% inhibition of the structurally related AGC kinases AKT, PKA, ROCK2, p70S6K, MSK, RSK1/2, and SGK. We demonstrate that AT13148 has antiproliferative activity in a range of in vitro models harboring relevant genetic abnormalities, including PTEN, KRAS, PIK3CA and HER2 aberrations. AT13148 caused substantial blockade of AKT, p70S6K, PKA, ROCK and SGK substrate phosphorylation and induction of apoptosis in both a concentration and time-dependent manner in cancer cells with clinically relevant genetic defects both in vitro and in vivo. Antitumor efficacy in HER2-positive, PIK3CA-mutant BT474 breast, PTEN-deficient PC3 human prostate cancer and PTEN-deficient MES-SA uterine tumor xenografts was demonstrated. We show for the first time that induction of AKT phosphorylation at serine 473 by AT13148, as reported for other ATP competitive inhibitors of AKT, is not a therapeutically relevant reactivation step for this compound. We used gene expression microarray studies to characterize the underlying molecular mechanisms of action of AT13148 and the selective AKT inhibitor CCT128930, and observed the induction of upstream regulators including insulin receptor substrate-2 and PIK3IP1 due to compensatory feedback loops, consistent with blockade of AKT signaling. These studies also showed that AT13148 and CCT128930 have distinct molecular effects in cancer cells: AT13148 had a predominant effect on apoptosis genes and caused a greater apoptotic phenotype, while CCT128930 modulated genes in the network regulating cell cycle. This finding emphasizes the functional differences of AT13148 as a multi-AGC kinase inhibitor in contrast to a more AKT-selective inhibitor. In view of the potential mechanistic advantages detailed above, and the potent antitumor activity observed at well tolerated doses against established human tumor xenografts with clinically relevant genetic drivers, the clinical utility of such an AGC kinase inhibitor strategy will now be assessed in a first-in-human Phase I trial of AT13148. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 928. doi:1538-7445.AM2012-928
Journal of Medicinal Chemistry | 2005
Adrian Liam Gill; Martyn Frederickson; Anne Cleasby; Steven John Woodhead; Maria Grazia Carr; Andrew James Woodhead; Margaret T. Walker; Miles Congreve; Lindsay A. Devine; Dominic Tisi; Marc O'Reilly; Lisa Seavers; Deborah J. Davis; Jayne Curry; Rachel Anthony; Alessandro Padova; Christopher W. Murray; Robin A. E. Carr; Harren Jhoti
Journal of Medicinal Chemistry | 2007
Gordon Saxty; Steven John Woodhead; Valerio Berdini; Thomas G. Davies; Marcel L. Verdonk; Paul G. Wyatt; Robert George Boyle; David Barford; Robert Downham; and Michelle D. Garrett; Robin A. E. Carr
Archive | 2005
Valerio Berdini; Robert George Boyle; Gordon Saxty; David Winter Walker; Steven John Woodhead; Paul Graham Wyatt; Alastair Donald; John Caldwell; Ian Collins; Tatiana Faria Da Fonseca
Journal of Molecular Biology | 2007
Thomas G. Davies; Marcel L. Verdonk; Brent Graham; Susanne Maria Saalau-Bethell; Christopher Charles Frederick Hamlett; Tatiana McHardy; Ian Collins; Michelle D. Garrett; Paul Workman; Steven John Woodhead; Harren Jhoti; David Barford
Archive | 2009
Gilbert Ebai Besong; Christopher Thomas Brain; Clinton A. Brooks; Miles Stuart Congreve; Claudio Dagostin; Guo He; Ying Hou; Steven Howard; Yue Li; Yipin Lu; Paul N. Mortenson; Troy Smith; Moo Sung; Steven John Woodhead; Wojciech Wrona