Network


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

Hotspot


Dive into the research topics where Robert Hugh Bradbury is active.

Publication


Featured researches published by Robert Hugh Bradbury.


Journal of Medicinal Chemistry | 2014

Discovery of a Potent and Selective EGFR Inhibitor (AZD9291) of Both Sensitizing and T790M Resistance Mutations That Spares the Wild Type Form of the Receptor

M. Raymond V. Finlay; Mark J. Anderton; Susan Ashton; Peter Ballard; Paul A. Bethel; Matthew R. Box; Robert Hugh Bradbury; Simon Brown; Sam Butterworth; Andrew Campbell; Christopher G. Chorley; Nicola Colclough; Darren Cross; Gordon S. Currie; Matthew Grist; Lorraine Hassall; George B. Hill; Daniel S. James; Michael James; Paul D. Kemmitt; Teresa Klinowska; Gillian M. Lamont; Scott Lamont; Nathaniel G. Martin; Heather L. McFarland; Martine J. Mellor; Jonathon P. Orme; David Perkins; Paula Perkins; Graham Richmond

Epidermal growth factor receptor (EGFR) inhibitors have been used clinically in the treatment of non-small-cell lung cancer (NSCLC) patients harboring sensitizing (or activating) mutations for a number of years. Despite encouraging clinical efficacy with these agents, in many patients resistance develops leading to disease progression. In most cases, this resistance is in the form of the T790M mutation. In addition, EGFR wild type receptor inhibition inherent with these agents can lead to dose limiting toxicities of rash and diarrhea. We describe herein the evolution of an early, mutant selective lead to the clinical candidate AZD9291, an irreversible inhibitor of both EGFR sensitizing (EGFRm+) and T790M resistance mutations with selectivity over the wild type form of the receptor. Following observations of significant tumor inhibition in preclinical models, the clinical candidate was administered clinically to patients with T790M positive EGFR-TKI resistant NSCLC and early efficacy has been observed, accompanied by an encouraging safety profile.


Cancer Research | 2016

AZD9496: An Oral Estrogen Receptor Inhibitor That Blocks the Growth of ER-Positive and ESR1-Mutant Breast Tumors in Preclinical Models.

Hazel M. Weir; Robert Hugh Bradbury; Mandy Lawson; Alfred A. Rabow; David Buttar; Rowena Callis; Jon Owen Curwen; Camila de Almeida; Peter Ballard; Micheal Hulse; Craig S. Donald; Lyman Feron; Galith Karoutchi; Philip A. MacFaul; Thomas A. Moss; Richard A. Norman; Stuart E. Pearson; Michael Tonge; Gareth Davies; Graeme Walker; Zena Wilson; Rachel Rowlinson; Steve Powell; Claire Sadler; Graham Richmond; Brendon Ladd; Ermira Pazolli; Anne Marie Mazzola; Celina D'Cruz; Chris De Savi

Fulvestrant is an estrogen receptor (ER) antagonist administered to breast cancer patients by monthly intramuscular injection. Given its present limitations of dosing and route of administration, a more flexible orally available compound has been sought to pursue the potential benefits of this drug in patients with advanced metastatic disease. Here we report the identification and characterization of AZD9496, a nonsteroidal small-molecule inhibitor of ERα, which is a potent and selective antagonist and downregulator of ERα in vitro and in vivo in ER-positive models of breast cancer. Significant tumor growth inhibition was observed as low as 0.5 mg/kg dose in the estrogen-dependent MCF-7 xenograft model, where this effect was accompanied by a dose-dependent decrease in PR protein levels, demonstrating potent antagonist activity. Combining AZD9496 with PI3K pathway and CDK4/6 inhibitors led to further growth-inhibitory effects compared with monotherapy alone. Tumor regressions were also seen in a long-term estrogen-deprived breast model, where significant downregulation of ERα protein was observed. AZD9496 bound and downregulated clinically relevant ESR1 mutants in vitro and inhibited tumor growth in an ESR1-mutant patient-derived xenograft model that included a D538G mutation. Collectively, the pharmacologic evidence showed that AZD9496 is an oral, nonsteroidal, selective estrogen receptor antagonist and downregulator in ER(+) breast cells that could provide meaningful benefit to ER(+) breast cancer patients. AZD9496 is currently being evaluated in a phase I clinical trial. Cancer Res; 76(11); 3307-18. ©2016 AACR.


Molecular Cancer Therapeutics | 2013

AZD3514: A Small Molecule That Modulates Androgen Receptor Signaling and Function In Vitro and In Vivo

Sarah A. Loddick; Sarah Ross; Andrew G. Thomason; David M. Robinson; Graeme Walker; Tom P.J. Dunkley; Sandra R. Brave; Nicola Broadbent; Natalie Stratton; Dawn Trueman; Elizabeth Mouchet; Fadhel Shaheen; Vivien Jacobs; Marie Cumberbatch; Joanne Wilson; Rhys D.O. Jones; Robert Hugh Bradbury; Alfred A. Rabow; Luke Gaughan; Chris Womack; Simon T. Barry; Craig N. Robson; Susan E. Critchlow; Stephen R. Wedge; A. Nigel Brooks

Continued androgen receptor (AR) expression and signaling is a key driver in castration-resistant prostate cancer (CRPC) after classical androgen ablation therapies have failed, and therefore remains a target for the treatment of progressive disease. Here, we describe the biological characterization of AZD3514, an orally bioavailable drug that inhibits androgen-dependent and -independent AR signaling. AZD3514 modulates AR signaling through two distinct mechanisms, an inhibition of ligand-driven nuclear translocation of AR and a downregulation of receptor levels, both of which were observed in vitro and in vivo. AZD3514 inhibited testosterone-driven seminal vesicle development in juvenile male rats and the growth of androgen-dependent Dunning R3327H prostate tumors in adult rats. Furthermore, this class of compound showed antitumor activity in the HID28 mouse model of CRPC in vivo. AZD3514 is currently in phase I clinical evaluation. Mol Cancer Ther; 12(9); 1715–27. ©2013 AACR.


Journal of Medicinal Chemistry | 2015

Optimization of a Novel Binding Motif to (E)-3-(3,5-Difluoro-4-((1R,3R)-2-(2-Fluoro-2-Methylpropyl)-3-Methyl-2, 3,4,9-Tetrahydro-1H-Pyrido[3,4-B]Indol-1-Yl)Phenyl)Acrylic Acid (Azd9496), a Potent and Orally Bioavailable Selective Estrogen Receptor Downregulator and Antagonist.

Chris De Savi; Robert Hugh Bradbury; Alfred A. Rabow; Richard A. Norman; Camila de Almeida; David M. Andrews; Peter Ballard; David Buttar; Rowena Callis; Gordon S. Currie; Jon Owen Curwen; Christopher D. Davies; Craig S. Donald; Lyman Feron; Helen Gingell; Steven C. Glossop; Barry R. Hayter; Syeed Hussain; Galith Karoutchi; Scott Lamont; Philip A. MacFaul; Thomas A. Moss; Stuart E. Pearson; Michael Tonge; Graeme Walker; Hazel M. Weir; Zena Wilson

The discovery of an orally bioavailable selective estrogen receptor downregulator (SERD) with equivalent potency and preclinical pharmacology to the intramuscular SERD fulvestrant is described. A directed screen identified the 1-aryl-2,3,4,9-tetrahydro-1H-pyrido[3,4-b]indole motif as a novel, druglike ER ligand. Aided by crystal structures of novel ligands bound to an ER construct, medicinal chemistry iterations led to (E)-3-(3,5-difluoro-4-((1R,3R)-2-(2-fluoro-2-methylpropyl)-3-methyl-2,3,4,9-tetrahydro-1H-pyrido[3,4-b]indol-1-yl)phenyl)acrylic acid (30b, AZD9496), a clinical candidate with high oral bioavailability across preclinical species that is currently being evaluated in phase I clinical trials for the treatment of advanced estrogen receptor (ER) positive breast cancer.


Bioorganic & Medicinal Chemistry Letters | 2011

Small-molecule androgen receptor downregulators as an approach to treatment of advanced prostate cancer

Robert Hugh Bradbury; Neil J. Hales; Alfred A. Rabow; Graeme Walker; David G. Acton; David M. Andrews; Peter Ballard; Nigel Brooks; Nicola Colclough; Alan Girdwood; Urs Hancox; Owen Jones; David Jude; Sarah A. Loddick; Andrew Austen Mortlock

Chemical starting points were investigated for downregulation of the androgen receptor as an approach to treatment of advanced prostate cancer. Although prototypic steroidal downregulators such as 6a designed for intramuscular administration showed insufficient cellular potency, a medicinal chemistry program derived from a novel androgen receptor ligand 8a led to 6-[4-(4-cyanobenzyl)piperazin-1-yl]-3-(trifluoromethyl)[1,2,4]triazolo[4,3-b]pyridazine (10b), for which high plasma levels following oral administration in a preclinical model compensate for moderate cellular potency.


Bioorganic & Medicinal Chemistry Letters | 2013

Discovery of AZD3514, a small-molecule androgen receptor downregulator for treatment of advanced prostate cancer.

Robert Hugh Bradbury; David G. Acton; Nicola Broadbent; A. Nigel Brooks; Gregory Richard Carr; Glenn Hatter; Barry R. Hayter; Kathryn Jane Hill; Nicholas J. Howe; Rhys D.O. Jones; David Jude; Scott Lamont; Sarah A. Loddick; Heather L. McFarland; Zaieda Parveen; Alfred A. Rabow; Gorkhn Sharma-Singh; Natalie Stratton; Andrew G. Thomason; Dawn Trueman; Graeme Walker; Stuart L. Wells; Joanne Wilson; J. Matthew Wood

Removal of the basic piperazine nitrogen atom, introduction of a solubilising end group and partial reduction of the triazolopyridazine moiety in the previously-described lead androgen receptor downregulator 6-[4-(4-cyanobenzyl)piperazin-1-yl]-3-(trifluoromethyl)[1,2,4]triazolo[4,3-b]pyridazine (1) addressed hERG and physical property issues, and led to clinical candidate 6-(4-{4-[2-(4-acetylpiperazin-1-yl)ethoxy]phenyl}piperidin-1-yl)-3-(trifluoromethyl)-7,8-dihydro[1,2,4]triazolo[4,3-b]pyridazine (12), designated AZD3514, that is being evaluated in a Phase I clinical trial in patients with castrate-resistant prostate cancer.


Cancer Research | 2016

The PARP Inhibitor AZD2461 Provides Insights into the Role of PARP3 Inhibition for Both Synthetic Lethality and Tolerability with Chemotherapy in Preclinical Models.

Lenka Oplustil O'Connor; Stuart L. Rulten; Aaron Cranston; Rajesh Odedra; Henry Brown; Janneke E. Jaspers; Louise Jones; Charlotte Knights; Bastiaan Evers; Attilla Ting; Robert Hugh Bradbury; Marina Pajic; Sven Rottenberg; Jos Jonkers; David Alan Rudge; Niall Morrison Barr Martin; Keith W. Caldecott; Alan Lau; Mark J. O'Connor

The PARP inhibitor AZD2461 was developed as a next-generation agent following olaparib, the first PARP inhibitor approved for cancer therapy. In BRCA1-deficient mouse models, olaparib resistance predominantly involves overexpression of P-glycoprotein, so AZD2461 was developed as a poor substrate for drug transporters. Here we demonstrate the efficacy of this compound against olaparib-resistant tumors that overexpress P-glycoprotein. In addition, AZD2461 was better tolerated in combination with chemotherapy than olaparib in mice, which suggests that AZD2461 could have significant advantages over olaparib in the clinic. However, this superior toxicity profile did not extend to rats. Investigations of this difference revealed a differential PARP3 inhibitory activity for each compound and a higher level of PARP3 expression in bone marrow cells from mice as compared with rats and humans. Our findings have implications for the use of mouse models to assess bone marrow toxicity for DNA-damaging agents and inhibitors of the DNA damage response. Finally, structural modeling of the PARP3-active site with different PARP inhibitors also highlights the potential to develop compounds with different PARP family member specificity profiles for optimal antitumor activity and tolerability. Cancer Res; 76(20); 6084-94. ©2016 AACR.


ACS Medicinal Chemistry Letters | 2013

Discovery of AZD8931, an Equipotent, Reversible Inhibitor of Signaling by EGFR, HER2, and HER3 Receptors.

Bernard Barlaam; Judith Anderton; Peter Ballard; Robert Hugh Bradbury; Laurent Francois Andre Hennequin; D. Mark Hickinson; Jason Grant Kettle; George Kirk; Teresa Klinowska; Christine Lambert-van der Brempt; Cath Trigwell; John Vincent; Donald J. Ogilvie

Deregulation of HER family signaling promotes proliferation and tumor cell survival and has been described in many human cancers. Simultaneous, equipotent inhibition of EGFR-, HER2-, and HER3-mediated signaling may be of clinical utility in cancer settings where the selective EGFR or HER2 therapeutic agents are ineffective or only modestly active. We describe the discovery of AZD8931 (2), an equipotent, reversible inhibitor of EGFR-, HER2-, and HER3-mediated signaling and the structure-activity relationships within this series. Docking studies based on a model of the HER2 kinase domain helped rationalize the increased HER2 activity seen with the methyl acetamide side chain present in AZD8931. AZD8931 exhibited good pharmacokinetics in preclinical species and showed superior activity in the LoVo tumor growth efficacy model compared to close analogues. AZD8931 is currently being evaluated in human clinical trials for the treatment of cancer.


Journal of The Chemical Society-perkin Transactions 1 | 1992

2-Amino ketene S,S-acetals as α-amino acid homoenolate equivalents. Synthesis of 3-substituted prolines and molecular structure of 2-(N-pivaloylpyrrolidin-2-ylidene)-1,3-dithiane

William O. Moss; Annette C. Jones; Richard Wisedale; Mary F. Mahon; Kieran C. Molloy; Robert Hugh Bradbury; Neil J. Hales; Timothy Gallagher

Allylic deprotonation of the heterocyclic 2-amino ketene S,S-acetal 8a, followed by regioselective γ-alkylation reaction of the resulting organolithium 10(a proline homoenolate equivalent) with electrophiles, leads to adduct 11. Controlled hydrolytic cleavage of 11 gives a series of 3-substituted prolines, including the conformationally-constrained aspartate and glutamate derivatives, 14e and 14f respectively. The bicyclic thiolactam 18 has been prepared in an attempt to provide an asymmetric variant of organolithium 10 but efforts to generate the requisite ketene N,S-acetal 19 were unsuccessful. Extension of the ketene S,S-acetal chemistry to other ring sizes has been examined within the context of substituted azetidine-2-carboxylates. Condensation of the protected amino ester 20 with AIMe3–HS(CH2)3SH was complicated, however, by the reactivity of the four-membered ring and led to the ring-opened adduct 24, with none of the required ketene S,S-acetal 22 being observed.


Journal of Medicinal Chemistry | 2016

Optimization of a Series of Bivalent Triazolopyridazine Based Bromodomain and Extraterminal Inhibitors: The Discovery of (3R)-4-[2-[4-[1-(3-Methoxy-[1,2,4]triazolo[4,3-b]pyridazin-6-yl)-4-piperidyl]phenoxy]ethyl]-1,3-dimethyl-piperazin-2-one (AZD5153)

Robert Hugh Bradbury; Rowena Callis; Gregory Richard Carr; Huawei Chen; Edwin Clark; Lyman Feron; Steve C. Glossop; Mark A. Graham; Maureen Hattersley; Chris Jones; Scott Lamont; Gilles Ouvry; Anil Patel; Joe Patel; Alfred A. Rabow; Craig A. Roberts; Stephen Stokes; Natalie Stratton; Graeme Walker; Lara Ward; David Whalley; David Whittaker; Gail Wrigley; Michael J. Waring

Here we report the discovery and optimization of a series of bivalent bromodomain and extraterminal inhibitors. Starting with the observation of BRD4 activity of compounds from a previous program, the compounds were optimized for BRD4 potency and physical properties. The optimized compound from this campaign exhibited excellent pharmacokinetic profile and exhibited high potency in vitro and in vivo effecting c-Myc downregulation and tumor growth inhibition in xenograft studies. This compound was selected as the development candidate, AZD5153. The series showed enhanced potency as a result of bivalent binding and a clear correlation between BRD4 activity and cellular potency.

Collaboration


Dive into the Robert Hugh Bradbury's collaboration.

Researchain Logo
Decentralizing Knowledge