Network


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

Hotspot


Dive into the research topics where Alexandra Stowell is active.

Publication


Featured researches published by Alexandra Stowell.


European Journal of Medicinal Chemistry | 2016

The discovery of 2-substituted phenol quinazolines as potent RET kinase inhibitors with improved KDR selectivity.

Rebecca Newton; Katherine A. Bowler; Emily M. Burns; Philip J. Chapman; Emma Fairweather; Samantha J R Fritzl; Kristin M. Goldberg; Niall M. Hamilton; Sarah Holt; Gemma Hopkins; Stuart Jones; Allan M. Jordan; Amanda J. Lyons; H. Nikki March; Neil Q. McDonald; Laura Maguire; Daniel Mould; Andrew Purkiss; Helen Small; Alexandra Stowell; Graeme J. Thomson; Ian Waddell; Bohdan Waszkowycz; Amanda J. Watson; Donald J. Ogilvie

Deregulation of the receptor tyrosine kinase RET has been implicated in medullary thyroid cancer, a small percentage of lung adenocarcinomas, endocrine-resistant breast cancer and pancreatic cancer. There are several clinically approved multi-kinase inhibitors that target RET as a secondary pharmacology but additional activities, most notably inhibition of KDR, lead to dose-limiting toxicities. There is, therefore, a clinical need for more specific RET kinase inhibitors. Herein we report our efforts towards identifying a potent and selective RET inhibitor using vandetanib 1 as the starting point for structure-based drug design. Phenolic anilinoquinazolines exemplified by 6 showed improved affinities towards RET but, unsurprisingly, suffered from high metabolic clearance. Efforts to mitigate the metabolic liability of the phenol led to the discovery that a flanking substituent not only improved the hepatocyte stability, but could also impart a significant gain in selectivity. This culminated in the identification of 36; a potent RET inhibitor with much improved selectivity against KDR.


Analytical Biochemistry | 2016

A high-throughput screening-compatible homogeneous time-resolved fluorescence assay measuring the glycohydrolase activity of human poly(ADP-ribose) glycohydrolase.

Alexandra Stowell; Dominic I. James; Ian Waddell; Neil Bennett; Caroline M. Truman; Ian Hardern; Donald J. Ogilvie

Poly(ADP-ribose) (PAR) polymers are transient post-translational modifications, and their formation is catalyzed by poly(ADP-ribose) polymerase (PARP) enzymes. A number of PARP inhibitors are in advanced clinical development for BRCA-mutated breast cancer, and olaparib has recently been approved for BRCA-mutant ovarian cancer; however, there has already been evidence of developed resistance mechanisms. Poly(ADP-ribose) glycohydrolase (PARG) catalyzes the hydrolysis of the endo- and exo-glycosidic bonds within the PAR polymers. As an alternative strategy, PARG is a potentially attractive therapeutic target. There is only one PARG gene, compared with 17 known PARP family members, and therefore a PARG inhibitor may have wider application with fewer compensatory mechanisms. Prior to the initiation of this project, there were no known existing cell-permeable small molecule PARG inhibitors for use as tool compounds to assess these hypotheses and no suitable high-throughput screening (HTS)-compatible biochemical assays available to identify start points for a drug discovery project. The development of this newly described high-throughput homogeneous time-resolved fluorescence (HTRF) assay has allowed HTS to proceed and, from this, the identification and advancement of multiple validated series of tool compounds for PARG inhibition.


F1000Research | 2016

Identification of selective inhibitors of RET and comparison with current clinical candidates through development and validation of a robust screening cascade

Amanda J. Watson; Gemma Hopkins; Samantha Hitchin; Habiba Begum; Stuart Jones; Allan M. Jordan; Sarah Holt; H. Nikki March; Rebecca Newton; Helen Small; Alexandra Stowell; Ian Waddell; Bohdan Waszkowycz; Donald J. Ogilvie

RET (REarranged during Transfection) is a receptor tyrosine kinase, which plays pivotal roles in regulating cell survival, differentiation, proliferation, migration and chemotaxis. Activation of RET is a mechanism of oncogenesis in medullary thyroid carcinomas where both germline and sporadic activating somatic mutations are prevalent. At present, there are no known specific RET inhibitors in clinical development, although many potent inhibitors of RET have been opportunistically identified through selectivity profiling of compounds initially designed to target other tyrosine kinases. Vandetanib and cabozantinib, both multi-kinase inhibitors with RET activity, are approved for use in medullary thyroid carcinoma, but additional pharmacological activities, most notably inhibition of vascular endothelial growth factor - VEGFR2 (KDR), lead to dose-limiting toxicity. The recent identification of RET fusions present in ~1% of lung adenocarcinoma patients has renewed interest in the identification and development of more selective RET inhibitors lacking the toxicities associated with the current treatments. In an earlier publication [Newton et al, 2016; 1] we reported the discovery of a series of 2-substituted phenol quinazolines as potent and selective RET kinase inhibitors. Here we describe the development of the robust screening cascade which allowed the identification and advancement of this chemical series. Furthermore we have profiled a panel of RET-active clinical compounds both to validate the cascade and to confirm that none display a RET-selective target profile.


Cancer Research | 2014

Abstract 2745: Pharmacological characterisation of cell active inhibitors of Poly(ADP-ribose) glycohydrolase (PARG)

Dominic I. James; Allan M. Jordan; Nicola Hamilton; Alison McGonagle; Kate Smith; Alexandra Stowell; Ian Waddell; Bohdan Waszkowycz; Donald J. Ogilvie

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA Poly(ADP-ribose) glycohydrolase (PARG) is the only enzyme known to catalyse hydrolysis of the O-glycosidic linkages of ADP-ribose polymers, thereby reversing the effects of poly(ADP-ribose) polymerases. Total PARG deficiency leads to cell death whilst PARG depletion, using RNAi, leads to pleiotropic effects such as PAR chain persistence, progression of single- to double-strand DNA lesions and NAD+ depletion. Whilst efforts to develop small molecule inhibitors of PARG activity have generally been hampered by poor physiochemical properties, off-target pharmacology and a lack of cell permeability, we have now developed a series of PARG inhibitors which have proved to be useful biological tool compounds. Displaying selective activity in both biochemical and, more importantly, cellular assays of PARG function, these derivatives have allowed an exploration of the phenotypes resulting from reversible, pharmacological PARG inhibition. Citation Format: Dominic James, Allan Jordan, Nicola Hamilton, Alison McGonagle, Kate Smith, Alexandra Stowell, Ian Waddell, Bohdan Waszkowycz, Donald Ogilvie. Pharmacological characterisation of cell active inhibitors of Poly(ADP-ribose) glycohydrolase (PARG). [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 2745. doi:10.1158/1538-7445.AM2014-2745


Molecular Cancer Therapeutics | 2015

Abstract C39: First-in-class inhibitors of the putatively undruggable DNA repair target Poly(ADP-ribose) glycohydrolase (PARG)

Bohdan Waszkowycz; Dominic I. James; Ben Acton; Emma Fairweather; Sam Fritzl; Niall M. Hamilton; Nicola Hamilton; Sarah Holt; James Hitchen; Colin Hutton; Stuart Donald Jones; Allan M. Jordan; Alison McGonagle; Daniel Mould; Helen Small; Kate Smith; Alexandra Stowell; Ian Waddell; Donald J. Ogilvie

Poly(ADP-ribose) glycohydrolase (PARG) is the only enzyme known to catalyse hydrolysis of the O-glycosidic linkages of ADP-ribose polymers, thereby reversing the effects of poly(ADP-ribose) polymerases (PARPs). PARG depletion, using RNAi, results in several effects such as PAR chain persistence, progression of single- to double-strand DNA lesions and NAD+ depletion. Given these findings, inhibition of PARG with a small molecule agent offers a potential opportunity to interfere with DNA repair mechanisms and induce cell death in those cells with increased susceptibility to DNA damage, such as tumour cells. Previous efforts to develop small molecule inhibitors of PARG activity have generally been hampered by poor physicochemical properties, off-target pharmacology and a lack of cell permeability, leading some to suggest that PARG may be undruggable. In contrast, we have now developed a series of first-in-class PARG inhibitors which display drug-like properties and attractive pharmacokinetic parameters. These compounds have proved to be useful biological tool compounds. Moreover, displaying selective activity in both biochemical and, more importantly, cellular assays of PARG function, these derivatives have allowed an exploration of the phenotypes resulting from reversible, pharmacological PARG inhibition in both in vitro cell panels and in vivo models. Furthermore, our initial bioinformatic analysis suggests that deficiency of a known tumour suppressor confers sensitivity to PARG inhibition, suggesting patient populations that will potentially benefit from PARGi therapies. Citation Format: Bohdan Waszkowycz, Dominic James, Ben Acton, Emma Fairweather, Sam Fritzl, Niall Hamilton, Nicola Hamilton, Sarah Holt, James Hitchen, Colin Hutton, Stuart Jones, Allan Jordan, Alison McGonagle, Daniel Mould, Helen Small, Kate Smith, Alexandra Stowell, Ian D. Waddell, Donald Ogilvie. First-in-class inhibitors of the putatively undruggable DNA repair target Poly(ADP-ribose) glycohydrolase (PARG). [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2015 Nov 5-9; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2015;14(12 Suppl 2):Abstract nr C39.


Cancer Research | 2015

Abstract 778: The identification and structure-guided optimisation of potent and selective inhibitors of oncogenes in medullary thyroid carcinoma and lung adenocarcinoma

Roger J. Butlin; Rebecca Newton; Mandy Watson; Gemma Hopkins; Ben Acton; Kate Bowler; Samantha J R Fritzl; Kristin M. Goldberg; Niall M. Hamilton; Sarah Holt; Stuart Jones; Allan M. Jordan; Nikki March; Daniel Mould; Helen Small; Alexandra Stowell; Ian Waddell; Bohdan Waszkowycz; Donald J. Ogilvie

Proceedings: AACR 106th Annual Meeting 2015; April 18-22, 2015; Philadelphia, PA RET (REarranged during Transfection) is a receptor tyrosine kinase (TK), which plays pivotal roles in regulating cell survival, differentiation, proliferation, migration and chemotaxis. Activating mutations in RET (C634W and M918T) have been identified in both familial and sporadic forms of medullary thyroid carcinoma (MTC) and correlate with aggressive disease progression, validating RET as a classical oncogene. Furthermore the recent identification of RET fusions (CCDC6-RET and KIF5B-RET) present in ∼1% of lung adenocarcinoma (LAD) patients has renewed interest in the identification and development of more selective RET inhibitors lacking the toxicities associated with the current treatments. At present, there are no known specific RET inhibitors in clinical development, although many potent inhibitors of RET have been identified opportunistically through selectivity profiling of compounds initially designed to target other TKs. Such “secondary RET inhibitors” include the clinical agents Vandetanib and Cabozantinib, both approved for use in MTC, but additional pharmacological activities (most notably inhibition of KDR) lead to dose-limiting toxicity. Using a robust screening cascade developed in house, we have measured RET and KDR inhibitory activity in vitro and in relevant cell line models to assess compound potency and selectivity. Anti-proliferative activity and off-target toxicity of these agents have also been measured. Although these competitor compounds displayed reasonable RET potency in cellular assays and this translated into anti-proliferative effects in our MTC and LAD disease models, as expected none met our target candidate criteria, clearly highlighting the need for therapeutic agents with improved selectivity. Guided by structure-based drug design, we have identified and optimised a novel series of potent and selective inhibitors of the RET kinase domain. These agents met our stringent criteria for enzyme and cell selectivity and, whilst potent in a RET-driven cell line, display little overt toxicity in a matched non-RET driven cell line. Herein, we describe the chemical optimisation of these agents and, using structural information, rationalise their improved selectivity. Citation Format: Roger J. Butlin, Rebecca Newton, Mandy Watson, Gemma Hopkins, Ben Acton, Kate Bowler, Samantha Fritzl, Kristin Goldberg, Niall Hamilton, Sarah Holt, Stuart Jones, Allan Jordan, Nikki March, Daniel Mould, Helen Small, Alexandra Stowell, Ian Waddell, Bohdan Waszkowycz, Donald Ogilvie. The identification and structure-guided optimisation of potent and selective inhibitors of oncogenes in medullary thyroid carcinoma and lung adenocarcinoma. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 778. doi:10.1158/1538-7445.AM2015-778


ACS Chemical Biology | 2016

First-in-Class Chemical Probes against Poly(ADP-ribose) Glycohydrolase (PARG) Inhibit DNA Repair with Differential Pharmacology to Olaparib

Dominic I. James; Kate Smith; Allan M. Jordan; Emma Fairweather; Louise A. Griffiths; Nicola S. Hamilton; James R. Hitchin; Colin Hutton; Stuart Jones; Paul Kelly; Alison McGonagle; Helen Small; Alexandra Stowell; Julie Tucker; Ian Waddell; Bohdan Waszkowycz; Donald J. Ogilvie


Bioorganic & Medicinal Chemistry Letters | 2016

Discovery of potent inhibitors of the lysophospholipase autotaxin.

Pritom Shah; Anne Cheasty; Caroline Foxton; Tony Raynham; Muddasar Farooq; Irene Farre Gutierrez; Aurore Lejeune; Michelle Pritchard; Andrew P. Turnbull; Leon Pang; Paul Owen; Susan M. Boyd; Alexandra Stowell; Allan M. Jordan; Niall M. Hamilton; James R. Hitchin; Martin Stockley; Ellen MacDonald; Mar Jimenez Quesada; Elisabeth Trivier; Jana Skeete; Huib Ovaa; Wouter H. Moolenaar; Hamish Ryder


Cancer Research | 2018

Abstract 2994: Discovery of selective, noncovalent small molecule inhibitors of DNMT1 as an alternative to traditional DNA hypomethylating agents

Melissa B. Pappalardi; Mark Cockerill; Jessica L. Handler; Alexandra Stowell; Kathryn Keenan; Christian S. Sherk; Elisabeth A. Minthorn; Charles F. McHugh; Charlotte Burt; Kristen Wong; David T. Fosbenner; Mehul Patel; Jacques Briand; Helai P. Mohammad; Lourdes Rueda; Andrew Benowitz; Rab K. Prinjha; Dirk A. Heerding; Ryan G. Kruger; Ali Raoof; Allan M. Jordan; Bryan W. King; Michael T. McCabe


Cancer Research | 2018

Abstract LB-068: Development of a screening cascade to identify selective small molecule inhibitors of DNMT1

Alexandra Stowell; Graeme Thomson; Mark Cockerill; Charlotte Burt; Emma Fairweather; Ian Waddell; Ali Raoof; Allan M. Jordan; Donald gilvie; Melissa B. Pappalardi; Juan I. Luengo; Ryan G. Kruger; Chris Carpenter

Collaboration


Dive into the Alexandra Stowell's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ian Waddell

University of Manchester

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Helen Small

University of Manchester

View shared research outputs
Top Co-Authors

Avatar

Sarah Holt

University of Manchester

View shared research outputs
Top Co-Authors

Avatar

Daniel Mould

University of Manchester

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Stuart Jones

University of Manchester

View shared research outputs
Researchain Logo
Decentralizing Knowledge