Network


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

Hotspot


Dive into the research topics where Steven Howard is active.

Publication


Featured researches published by Steven Howard.


Journal of Medicinal Chemistry | 2009

Fragment-Based Discovery of the Pyrazol-4-Yl Urea (at9283), a Multitargeted Kinase Inhibitor with Potent Aurora Kinase Activity.

Steven Howard; Valerio Berdini; John A. Boulstridge; Maria Grazia Carr; David M. Cross; Jayne Curry; Lindsay A. Devine; Theresa Rachel Early; Lynsey Fazal; Adrian Liam Gill; Michelle Heathcote; Sarita Maman; Julia E. Matthews; Rachel McMenamin; Eva Figueroa Navarro; Michael A. O’Brien; Marc O’Reilly; David C. Rees; Matthias Reule; Dominic Tisi; Glyn Williams; Mladen Vinkovic; Paul G. Wyatt

Here, we describe the identification of a clinical candidate via structure-based optimization of a ligand efficient pyrazole-benzimidazole fragment. Aurora kinases play a key role in the regulation of mitosis and in recent years have become attractive targets for the treatment of cancer. X-ray crystallographic structures were generated using a novel soakable form of Aurora A and were used to drive the optimization toward potent (IC(50) approximately 3 nM) dual Aurora A/Aurora B inhibitors. These compounds inhibited growth and survival of HCT116 cells and produced the polyploid cellular phenotype typically associated with Aurora B kinase inhibition. Optimization of cellular activity and physicochemical properties ultimately led to the identification of compound 16 (AT9283). In addition to Aurora A and Aurora B, compound 16 was also found to inhibit a number of other kinases including JAK2 and Abl (T315I). This compound demonstrated in vivo efficacy in mouse xenograft models and is currently under evaluation in phase I clinical trials.


ACS Medicinal Chemistry Letters | 2012

Fragment-Based Discovery of 7-Azabenzimidazoles as Potent, Highly Selective, and Orally Active CDK4/6 Inhibitors

Young Shin Cho; Hayley Angove; Christopher Thomas Brain; Christine Hiu-Tung Chen; Hong Cheng; Robert Cheng; Rajiv Chopra; Kristy Chung; Miles Congreve; Claudio Dagostin; Deborah J. Davis; Ruth Feltell; John William Giraldes; Steven Douglas Hiscock; Sunkyu Kim; Steven Kovats; Bharat Lagu; Kim Lewry; Alice Loo; Yipin Lu; Michael Luzzio; Wiesia Maniara; Rachel McMenamin; Paul N. Mortenson; Rajdeep Kaur Benning; Marc O'Reilly; David C. Rees; Junqing Shen; Troy Smith; Yaping Wang

Herein, we describe the discovery of potent and highly selective inhibitors of both CDK4 and CDK6 via structure-guided optimization of a fragment-based screening hit. CDK6 X-ray crystallography and pharmacokinetic data steered efforts in identifying compound 6, which showed >1000-fold selectivity for CDK4 over CDKs 1 and 2 in an enzymatic assay. Furthermore, 6 demonstrated in vivo inhibition of pRb-phosphorylation and oral efficacy in a Jeko-1 mouse xenograft model.


ACS Medicinal Chemistry Letters | 2015

Fragment-Based Discovery of Type I Inhibitors of Maternal Embryonic Leucine Zipper Kinase

Christopher N. Johnson; Valerio Berdini; Lijs Beke; Pascal Bonnet; Dirk Brehmer; Joseph E. Coyle; Phillip J. Day; Martyn Frederickson; Eddy Jean Edgard Freyne; Ron Gilissen; Christopher Charles Frederick Hamlett; Steven Howard; Lieven Meerpoel; Rachel McMenamin; Sahil Patel; David C. Rees; Andrew Sharff; Francois Maria Sommen; Tongfei Wu; Joannes Theodorus Maria Linders

Fragment-based drug design was successfully applied to maternal embryonic leucine zipper kinase (MELK). A low affinity (160 μM) fragment hit was identified, which bound to the hinge region with an atypical binding mode, and this was optimized using structure-based design into a low-nanomolar and cell-penetrant inhibitor, with a good selectivity profile, suitable for use as a chemical probe for elucidation of MELK biology.


ACS Medicinal Chemistry Letters | 2015

Structure-Based Design of Type II Inhibitors Applied to Maternal Embryonic Leucine Zipper Kinase

Christopher Norbert Johnson; Christophe Denis Adelinet; Valerio Berdini; Lijs Beke; Pascal Bonnet; Dirk Brehmer; Frederick Calo; Joseph E. Coyle; Phillip J. Day; Martyn Frederickson; Eddy Jean Edgard Freyne; Ron Gilissen; Christopher Charles Frederick Hamlett; Steven Howard; Lieven Meerpoel; Laurence Anne Mevellec; Rachel McMenamin; Elisabeth Thérèse Jeanne Pasquier; Sahil Patel; David C. Rees; Joannes Theodorus Maria Linders

A novel Type II kinase inhibitor chemotype has been identified for maternal embryonic leucine zipper kinase (MELK) using structure-based ligand design. The strategy involved structural characterization of an induced DFG-out pocket by protein-ligand X-ray crystallography and incorporation of a slender linkage capable of bypassing a large gate-keeper residue, thus enabling design of molecules accessing both hinge and induced pocket regions. Optimization of an initial hit led to the identification of a low-nanomolar, cell-penetrant Type II inhibitor suitable for use as a chemical probe for MELK.


ACS Medicinal Chemistry Letters | 2013

Fragment-based discovery of 6-azaindazoles as inhibitors of bacterial DNA ligase.

Steven Howard; Nader Amin; Andrew Benowitz; Elisabetta Chiarparin; Haifeng Cui; Xiaodong Deng; Tom D. Heightman; David J. Holmes; Anna Hopkins; Jianzhong Huang; Qi Jin; Constantine Kreatsoulas; Agnes C. L. Martin; Frances Massey; Lynn McCloskey; Paul N. Mortenson; Puja Pathuri; Dominic Tisi; Pamela A. Williams

Herein we describe the application of fragment-based drug design to bacterial DNA ligase. X-ray crystallography was used to guide structure-based optimization of a fragment-screening hit to give novel, nanomolar, AMP-competitive inhibitors. The lead compound 13 showed antibacterial activity across a range of pathogens. Data to demonstrate mode of action was provided using a strain of S. aureus, engineered to overexpress DNA ligase.


Essays in Biochemistry | 2017

Fragment-based drug discovery and its application to challenging drug targets

Amanda J. Price; Steven Howard; Benjamin David Cons

Fragment-based drug discovery (FBDD) is a technique for identifying low molecular weight chemical starting points for drug discovery. Since its inception 20 years ago, FBDD has grown in popularity to the point where it is now an established technique in industry and academia. The approach involves the biophysical screening of proteins against collections of low molecular weight compounds (fragments). Although fragments bind to proteins with relatively low affinity, they form efficient, high quality binding interactions with the protein architecture as they have to overcome a significant entropy barrier to bind. Of the biophysical methods available for fragment screening, X-ray protein crystallography is one of the most sensitive and least prone to false positives. It also provides detailed structural information of the protein-fragment complex at the atomic level. Fragment-based screening using X-ray crystallography is therefore an efficient method for identifying binding hotspots on proteins, which can then be exploited by chemists and biologists for the discovery of new drugs. The use of FBDD is illustrated here with a recently published case study of a drug discovery programme targeting the challenging protein-protein interaction Kelch-like ECH-associated protein 1:nuclear factor erythroid 2-related factor 2.


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 | 2006

Application of Fragment Screening and Fragment Linking to the Discovery of Novel Thrombin Inhibitors

Nigel I. Howard; Chris Abell; Wendy Blakemore; Gianni Chessari; Miles Congreve; Steven Howard; Harren Jhoti; Christopher W. Murray; and Lisa C. A. Seavers; Rob L. M. van Montfort


Archive | 2004

Benzimidazole derivatives and their use as protein kinases inhibitors

Valerio Berdini; Michael Alistair O'brien; Maria Grazia Carr; Theresa Rachel Early; Eva Figueroa Navarro; Adrian Liam Gill; Steven Howard; Gary Trewartha; Alison Jo-Ann Woolford; Andrew James Woodhead; Paul Graham Wyatt


Archive | 2009

Pyrrolopyrimidine compounds as cdk inhibitors

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

Researchain Logo
Decentralizing Knowledge