Network


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

Hotspot


Dive into the research topics where James E. H. Day is active.

Publication


Featured researches published by James E. H. Day.


Journal of Medicinal Chemistry | 2015

Fragment-Based Drug Discovery Targeting Inhibitor of Apoptosis Proteins: Discovery of a Non-Alanine Lead Series with Dual Activity Against cIAP1 and XIAP.

Gianni Chessari; Ildiko Maria Buck; James E. H. Day; Philip J. Day; Aman Iqbal; Christopher Norbert Johnson; Edward J. Lewis; Vanessa Martins; Darcey Miller; Michael Reader; David C. Rees; Sharna J. Rich; Emiliano Tamanini; Marc Vitorino; George Ward; Pamela A. Williams; Glyn Williams; Nicola E. Wilsher; Alison Jo-Anne Woolford

Inhibitor of apoptosis proteins (IAPs) are important regulators of apoptosis and pro-survival signaling pathways whose deregulation is often associated with tumor genesis and tumor growth. IAPs have been proposed as targets for anticancer therapy, and a number of peptidomimetic IAP antagonists have entered clinical trials. Using our fragment-based screening approach, we identified nonpeptidic fragments binding with millimolar affinities to both cellular inhibitor of apoptosis protein 1 (cIAP1) and X-linked inhibitor of apoptosis protein (XIAP). Structure-based hit optimization together with an analysis of protein-ligand electrostatic potential complementarity allowed us to significantly increase binding affinity of the starting hits. Subsequent optimization gave a potent nonalanine IAP antagonist structurally distinct from all IAP antagonists previously reported. The lead compound had activity in cell-based assays and in a mouse xenograft efficacy model and represents a highly promising start point for further optimization.


Journal of Medicinal Chemistry | 2017

Discovery of a Potent Nonpeptidomimetic, Small-Molecule Antagonist of Cellular Inhibitor of Apoptosis Protein 1 (cIAP1) and X-Linked Inhibitor of Apoptosis Protein (XIAP).

Emiliano Tamanini; Ildiko Maria Buck; Gianni Chessari; Elisabetta Chiarparin; James E. H. Day; Martyn Frederickson; Charlotte Mary Griffiths-Jones; Keisha Hearn; Tom D. Heightman; Aman Iqbal; Christopher N. Johnson; Edward J. Lewis; Vanessa Martins; Torren Peakman; Michael Reader; Sharna J. Rich; George Ward; Pamela A. Williams; Nicola E. Wilsher

XIAP and cIAP1 are members of the inhibitor of apoptosis protein (IAP) family and are key regulators of anti-apoptotic and pro-survival signaling pathways. Overexpression of IAPs occurs in various cancers and has been associated with tumor progression and resistance to treatment. Structure-based drug design (SBDD) guided by structural information from X-ray crystallography, computational studies, and NMR solution conformational analysis was successfully applied to a fragment-derived lead resulting in AT-IAP, a potent, orally bioavailable, dual antagonist of XIAP and cIAP1 and a structurally novel chemical probe for IAP biology.


Cancer Research | 2013

Abstract 2944: AT-IAP, a dual cIAP1 and XIAP antagonist with oral antitumor activity in melanoma models.

Gianni Chessari; Ahn Maria; Ildiko Maria Buck; Elisabetta Chiarparin; Joe Coyle; James E. H. Day; Martyn Frederickson; Charlotte Mary Griffiths-Jones; Keisha Hearn; Steven Howard; Tom D. Heightman; Petra Hillmann; Aman Iqbal; Christopher N. Johnson; Jon Lewis; Vanessa Martins; Joanne M. Munck; Mike Reader; Lee Page; Anna Hopkins; Alessia Millemaggi; Caroline Richardson; Gordon Saxty; Tomoko Smyth; Emiliano Tamanini; Neil Thompson; George Ward; Glyn Williams; Pamela A. Williams; Nicola E. Wilsher

Melanoma is a highly aggressive malignancy with an exceptional ability to develop resistance and no curative therapy is available for patients with metastatic disease. Inhibitor of apoptosis proteins (IAP) play a key role in preventing cell death by apoptosis. In normal cell, IAPs are highly regulated by endogenous antagonists (e.g. SMAC) but in melanoma cell lines and in patient samples expression levels of IAPs are generally high and depleting IAPs by siRNA tended to reduce cell viability, with XIAP reduction being the most efficient [1]. Small molecule IAP antagonists have the ability to switch IAP-controlled pro-survival pathways towards apoptosis and cell death. Recent evidence suggests that a true dual antagonist of both cIAP1 and XIAP will promote an effective apoptotic response through generation of death-inducing ripoptosome complexes, with resultant caspase activation [2, 3]. We have used our fragment-based drug discovery technology PyramidTM to derive a non-peptidomimetic IAP antagonist, AT-IAP, which does not have an alanine warhead and has nanomolar cellular potency for both XIAP and cIAP1. Initial pharmacokinetic and pharmacodynamic modeling of AT-IAP in mice bearing the MDA-MB-231 cell line indicated that daily oral dosing of AT-IAP at 30 mg/kg ensures high concentrations of compound in tumor and plasma over a 24 h period with resultant inhibition of both XIAP and cIAP1 and induction of apoptosis markers (cleaved PARP and cleaved caspase-3). In this paper, we describe the characterization of AT-IAP in melanoma models. An in vitro cell line proliferation screen demonstrated that 36% of melanoma cell lines exhibited enhanced sensitivity to AT-IAP, which was improved on addition of exogenous 1 ng/ml TNF-α (92% of cell lines were sensitive to AT-IAP + TNF-α). Sensitivity of melanoma cells to AT-IAP has also been confirmed in a panel of 20 primary melanoma tumors in colony formation assays set up in the presence and absence of added TNF-α. Finally, a set of biomarkers has been identified and used to predict single agent activity of AT-IAP in a range of melanoma cell line and patient derived xenograft models. [1] Engesaeter et al., Cancer Biology & Therapy, 2011, 12 (1), 47 [2] Ndubaku et al., ACS Chem Biol., 2009, 4 (7), 557 [3] Meier, P., Nat Rev. Cancer, 2010, 10 (8), 561 Citation Format: Gianni Chessari, Ahn Maria, Ildiko Buck, Elisabetta Chiarparin, Joe Coyle, James Day, Martyn Frederickson, Charlotte Griffiths-Jones, Keisha Hearn, Steven Howard, Tom Heightman, Petra Hillmann, Aman Iqbal, Christopher N. Johnson, Jon Lewis, Vanessa Martins, Joanne Munck, Mike Reader, Lee Page, Anna Hopkins, Alessia Millemaggi, Caroline Richardson, Gordon Saxty, Tomoko Smyth, Emiliano Tamanini, Neil Thompson, George Ward, Glyn Williams, Pamela Williams, Nicola Wilsher, Alison Woolford. AT-IAP, a dual cIAP1 and XIAP antagonist with oral antitumor activity in melanoma models. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 2944. doi:10.1158/1538-7445.AM2013-2944


Journal of Medicinal Chemistry | 2018

Fragment-Based Discovery of a Potent, Orally Bioavailable Inhibitor That Modulates the Phosphorylation and Catalytic Activity of ERK1/2.

Tom D. Heightman; Valerio Berdini; Hannah Braithwaite; Ildiko Maria Buck; Megan Cassidy; Juan Castro; Aurélie Courtin; James E. H. Day; Charlotte East; Lynsey Fazal; Brent Graham; Charlotte Mary Griffiths-Jones; John Lyons; Vanessa Martins; Sandra Muench; Joanne M. Munck; David Norton; Marc O’Reilly; Nick Palmer; Puja Pathuri; Michael Reader; David C. Rees; Sharna J. Rich; Caroline Richardson; Harpreet K. Saini; Neil Thompson; Nicola G. Wallis; Hugh Walton; Nicola E. Wilsher; Alison Jo-Anne Woolford

Aberrant activation of the MAPK pathway drives cell proliferation in multiple cancers. Inhibitors of BRAF and MEK kinases are approved for the treatment of BRAF mutant melanoma, but resistance frequently emerges, often mediated by increased signaling through ERK1/2. Here, we describe the fragment-based generation of ERK1/2 inhibitors that block catalytic phosphorylation of downstream substrates such as RSK but also modulate phosphorylation of ERK1/2 by MEK without directly inhibiting MEK. X-ray crystallographic and biophysical fragment screening followed by structure-guided optimization and growth from the hinge into a pocket proximal to the C-α helix afforded highly potent ERK1/2 inhibitors with excellent kinome selectivity. In BRAF mutant cells, the lead compound suppresses pRSK and pERK levels and inhibits proliferation at low nanomolar concentrations. The lead exhibits tumor regression upon oral dosing in BRAF mutant xenograft models, providing a promising basis for further optimization toward clinical pERK1/2 modulating ERK1/2 inhibitors.


Cancer Research | 2012

Abstract 2018: Discovery of potent dual inhibitors of both XIAP and cIAP1 using fragment based drug discovery

Gianni Chessari; Ildiko Maria Buck; Elisabetta Chiarparin; James E. H. Day; Martyn Frederickson; Keisha Hearn; Tom D. Heightman; Petra Hillmann; Aman Iqbal; Christopher N. Johnson; Jon Lewis; Vanessa Martins; Caroline Richardson; Tomoko Smyth; Emiliano Tamanini; Neil Thompson; George Ward; Glyn Williams; Pamela A. Williams; Nicola E. Wilsher; Alison Jo-Anne Woolford

Proceedings: AACR 103rd Annual Meeting 2012‐‐ Mar 31‐Apr 4, 2012; Chicago, IL XIAP and cIAP1 are members of the inhibitor of apoptosis (IAP) protein family. Both proteins have the ability to attenuate apoptosis induced through intrinsic and extrinsic stimuli via inhibition of caspase-3, -7, -8 and -9. The defining feature of both XIAP and cIAP1 is the presence in their protein sequence of 3 Baculoviral IAP Repeat (BIR) domains, which are necessary for their antiapoptotic activity. The mitochondrial protein SMAC uses its N-terminal region (AVPI) to interact with BIR domains and deactivate the antiapoptotic function of IAPs. Several companies and academic groups have active programs developing SMAC peptidomimetic compounds based on the AVPI motif. In general, those compounds have the tendency to be cIAP1 selective like their tetrapeptide progenitor (AVPI IC50 values for XIAP-BIR3 and cIAP1-BIR3 are 0.3 uM and 0.016 uM respectively). Using our fragment-based screening approach, PyramidTM, we identified a non-peptidomimetic chemotype which binds with similar potency to the BIR3 domain of both XIAP and cIAP1. Hit optimisation using a structure based approach led to the discovery of potent true dual XIAP and cIAP1 antagonists with good in vivo physico-chemical profile and no P450 or hERG liabilities. Dual XIAP/cIAP1 inhibitors have potential for more effective apoptosis and less toxicity associated with cytokine production. Compounds were initially characterised in fluorescence polarisation binding assays using XIAP-BIR3 or cIAP1-BIR3 domains. Robust induction of apoptosis was observed in two sensitive breast cancer cell lines (EC50s well below 0.1 uM in EVSA-T and MDA-MB-231); whilst HCT116 cells (colon cancer) were insensitive (unless exogenous TNF-α was added). This in vitro cell line killing was demonstrated to correlate closely with cIAP1 antagonism and hence a parallel cell assay was established to measure XIAP antagonism. An engineered HEK293 cell line was stably co-transfected with full length FLAG-tagged human XIAP cDNA and full length (untagged) human caspase-9 cDNA. Inhibition of caspase-9 binding to XIAP was measured in immunoprecipitation assays. This gave us a sensitive read-out for XIAP antagonism in cells which could be plotted against the most sensitive cell killing read-out (from the EVSA-T cell line) to establish relative XIAP vs cIAP1 selectivities and to select dual antagonists of both IAPs. Potent compounds (HEK293-EC50 <0.01 uM and EVSA-T-EC50 <0.01 uM) were further characterised in PKPD studies in mice bearing MDA-MB-231 xenografts. Compounds with good oral exposure achieved high concentration in tumor over 24h periods which ensured excellent inhibition of both XIAP and cIAP1 with consequent reduction of cIAP1 levels and induction of apoptosis markers (PARP, Caspase-3). Finally, dual XIAP/cIAP1 inhibitors have been investigated in xenograft models (melanoma, breast and colorectal cancer) and have achieved significant efficacy at tolerated doses. 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 2018. doi:1538-7445.AM2012-2018


ACS Medicinal Chemistry Letters | 2015

Fragment-Based Discovery of Potent and Selective DDR1/2 Inhibitors

Christopher W. Murray; Valerio Berdini; Ildiko Maria Buck; Maria E. Carr; Anne Cleasby; Joseph E. Coyle; Jayne Curry; James E. H. Day; Phillip J. Day; Keisha Hearn; Aman Iqbal; Lydia Y. W. Lee; Vanessa Martins; Paul N. Mortenson; Joanne M. Munck; Lee W. Page; Sahil Patel; Susan Roomans; Kirsten Smith; Emiliano Tamanini; Gordon Saxty


Archive | 2012

Bicyclic heterocycle compounds and their uses in therapy

Alison Jo-Anne Woolford; Steven Howard; Ildiko Maria Buck; Gianni Chessari; Christopher N. Johnson; Emiliano Tamanini; James E. H. Day; Elisabetta Chiarparin; Thomas Daniel Heightman; Martyn Frederickson; Charlotte Mary Griffiths-Jones


Synlett | 2015

The Synthesis of 3,3-Dimethyl Aza- and Diazaindolines Using a Palladium-Catalysed Intramolecular Reductive Cyclisation

James E. H. Day; Martyn Frederickson; Colin Hogg; Christopher N. Johnson; Alistair Meek; Julian Northern; Michael Reader; Gary Reid


Molecular Cancer Therapeutics | 2018

Abstract B161: Fragment-based discovery of a highly potent, orally bioavailable ERK1/2 inhibitor that modulates the phosphorylation and catalytic activity of ERK1/2

Tom D. Heightman; Valerio Berdini; Hannah Braithwaite; Ildiko Maria Buck; Megan Cassidy; Juan Castro; Aurélie Courtin; James E. H. Day; Charlotte East; Lynsey Fazal; Brent Graham; Charlotte Mary Griffiths-Jones; John Lyons; Vanessa Martins; Sandra Muench; Joanne M. Munck; David Norton; Marc O'Reilly; Nick Palmer; Puja Pathuri; Mike Reader; David C. Rees; Sharna J. Rich; Caroline Richardson; Harpreet K. Saini; Neil Thomas Thompson; Nicola G. Wallis; Hugh Walton; Nicola E. Wilsher; Alison Jo-Anne Woolford


Molecular Cancer Therapeutics | 2018

Abstract B154: Characterization of a novel ERK1/2 inhibitor, which modulates the phosphorylation and catalytic activity of ERK1/2

Joanne M. Munck; Valerio Berdini; Luke Bevan; Hannah Braithwaite; Ildiko Maria Buck; Megan Cassidy; Juan Castro; Aurélie Courtin; James E. H. Day; Charlotte East; Lynsey Fazal; Brent Graham; Charlotte Mary Griffiths-Jones; Tom D. Heightman; Chris J. Hindley; Birikiti Kidane; Justyna Kucia-Tran; John Lyons; Vanessa Martins; Sandra Muench; Chris Murray; David Norton; Marc O'Reilly; Nick Palmer; Puja Pathuri; Mike Reader; David C. Rees; Sharna J. Rich; Caroline Richardson; Harpreet K. Saini

Researchain Logo
Decentralizing Knowledge