Thomas G. Davies
Astex
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Thomas G. Davies.
PLOS ONE | 2014
Anne Cleasby; Jeff Yon; Philip J. Day; Caroline Richardson; Ian J. Tickle; Pamela A. Williams; James F. Callahan; Robin Arthur Ellis Carr; Nestor O. Concha; Jeffrey K. Kerns; Hongwei Qi; Thomas D. Sweitzer; Paris Ward; Thomas G. Davies
The protein Keap1 is central to the regulation of the Nrf2-mediated cytoprotective response, and is increasingly recognized as an important target for therapeutic intervention in a range of diseases involving excessive oxidative stress and inflammation. The BTB domain of Keap1 plays key roles in sensing environmental electrophiles and in mediating interactions with the Cul3/Rbx1 E3 ubiquitin ligase system, and is believed to be the target for several small molecule covalent activators of the Nrf2 pathway. However, despite structural information being available for several BTB domains from related proteins, there have been no reported crystal structures of Keap1 BTB, and this has precluded a detailed understanding of its mechanism of action and interaction with antagonists. We report here the first structure of the BTB domain of Keap1, which is thought to contain the key cysteine residue responsible for interaction with electrophiles, as well as structures of the covalent complex with the antagonist CDDO/bardoxolone, and of the constitutively inactive C151W BTB mutant. In addition to providing the first structural confirmation of antagonist binding to Keap1 BTB, we also present biochemical evidence that adduction of Cys 151 by CDDO is capable of inhibiting the binding of Cul3 to Keap1, and discuss how this class of compound might exert Nrf2 activation through disruption of the BTB-Cul3 interface.
Journal of Medicinal Chemistry | 2008
John Caldwell; Thomas G. Davies; Alastair Donald; Tatiana McHardy; Martin G. Rowlands; G. W Aherne; Lisa-Jane K. Hunter; Kathryn R. Taylor; Ruth Ruddle; Florence I. Raynaud; Marinus Leendert Verdonk; Paul Workman; Garrett; Ian Collins
Fragment-based screening identified 7-azaindole as a protein kinase B inhibitor scaffold. Fragment elaboration using iterative crystallography of inhibitor-PKA-PKB chimera complexes efficiently guided improvements in the potency and selectivity of the compounds, resulting in the identification of nanomolar 6-(piperidin-1-yl)purine, 4-(piperidin-1-yl)-7-azaindole, and 4-(piperidin-1-yl)pyrrolo[2,3- d]pyrimidine inhibitors of PKBbeta with antiproliferative activity and showing pathway inhibition in cells. A divergence in the binding mode was seen between 4-aminomethylpiperidine and 4-aminopiperidine containing molecules. Selectivity for PKB vs PKA was observed with 4-aminopiperidine derivatives, and the most PKB-selective inhibitor (30-fold) showed significantly different bound conformations between PKA and PKA-PKB chimera.
Topics in Current Chemistry | 2011
Thomas G. Davies; Ian J. Tickle
The fragment-based approach is now well established as an important component of modern drug discovery. A key part in establishing its position as a viable technique has been the development of a range of biophysical methodologies with sufficient sensitivity to detect the binding of very weakly binding molecules. X-ray crystallography was one of the first techniques demonstrated to be capable of detecting such weak binding, but historically its potential for screening was under-appreciated and impractical due to its relatively low throughput. In this chapter we discuss the various benefits associated with fragment-screening by X-ray crystallography, and describe the technical developments we have implemented to allow its routine use in drug discovery. We emphasize how this approach has allowed a much greater exploitation of crystallography than has traditionally been the case within the pharmaceutical industry, with the rapid and timely provision of structural information having maximum impact on project direction.
Journal of Medicinal Chemistry | 2010
Tatiana McHardy; John Caldwell; Kwai-Ming Cheung; Lisa J. Hunter; Kevin Taylor; Martin G. Rowlands; Ruth Ruddle; Alan T. Henley; Alexis de Haven Brandon; Melanie Valenti; Thomas G. Davies; Lynsey Fazal; Lisa Seavers; Florence I. Raynaud; Suzanne A. Eccles; G. Wynne Aherne; Michelle D. Garrett; Ian Collins
Protein kinase B (PKB or Akt) is an important component of intracellular signaling pathways regulating growth and survival. Signaling through PKB is frequently deregulated in cancer, and inhibitors of PKB therefore have potential as antitumor agents. The optimization of lipophilic substitution within a series of 4-benzyl-1-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)piperidin-4-amines provided ATP-competitive, nanomolar inhibitors with up to 150-fold selectivity for inhibition of PKB over the closely related kinase PKA. Although active in cellular assays, compounds containing 4-amino-4-benzylpiperidines underwent metabolism in vivo, leading to rapid clearance and low oral bioavailability. Variation of the linker group between the piperidine and the lipophilic substituent identified 4-amino-1-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)piperidine-4-carboxamides as potent and orally bioavailable inhibitors of PKB. Representative compounds modulated biomarkers of signaling through PKB in vivo and strongly inhibited the growth of human tumor xenografts in nude mice at well-tolerated doses.
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 | 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 | 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
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