Network


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

Hotspot


Dive into the research topics where Alfred A. Rabow is active.

Publication


Featured researches published by Alfred A. Rabow.


Proteins | 2005

Linking tumor cell cytotoxicity to mechanism of drug action: an integrated analysis of gene expression, small-molecule screening and structural databases.

David G. Covell; Anders Wallqvist; Ruili Huang; Narmada Thanki; Alfred A. Rabow; Xiang-Jun Lu

An integrated, bioinformatic analysis of three databases comprising tumor‐cell‐based small molecule screening data, gene expression measurements, and PDB (Protein Data Bank) ligand–target structures has been developed for probing mechanism of drug action (MOA). Clustering analysis of GI50 profiles for the NCIs database of compounds screened across a panel of tumor cells (NCI60) was used to select a subset of unique cytotoxic responses for about 4000 small molecules. Drug–gene–PDB relationships for this test set were examined by correlative analysis of cytotoxic response and differential gene expression profiles within the NCI60 and structural comparisons with known ligand–target crystallographic complexes. A survey of molecular features within these compounds finds thirteen conserved Compound Classes, each class exhibiting chemical features important for interactions with a variety of biological targets. Protein targets for an additional twelve Compound Classes could be directly assigned using drug‐protein interactions observed in the crystallographic database. Results from the analysis of constitutive gene expressions established a clear connection between chemo‐resistance and overexpression of gene families associated with the extracellular matrix, cytoskeletal organization, and xenobiotic metabolism. Conversely, chemo‐sensitivity implicated overexpression of gene families involved in homeostatic functions of nucleic acid repair, aryl hydrocarbon metabolism, heat shock response, proteasome degradation and apoptosis. Correlations between chemo‐responsiveness and differential gene expressions identified chemotypes with nonselective (i.e., many) molecular targets from those likely to have selective (i.e., few) molecular targets. Applications of data mining strategies that jointly utilize tumor cell screening, genomic, and structural data are presented for hypotheses generation and identifying novel anticancer candidates. Proteins 2005. Published 2005 Wiley‐Liss, Inc.


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.


Nature Chemical Biology | 2016

Potent and selective bivalent inhibitors of BET bromodomains

Michael J. Waring; Huawei Chen; Alfred A. Rabow; Graeme Walker; Romel Bobby; Scott Boiko; Rob H. Bradbury; Rowena Callis; Edwin Clark; Ian L. Dale; Danette L. Daniels; Austin Dulak; Liz Flavell; Geoff Holdgate; Thomas A. Jowitt; Alexey Kikhney; Mark S. McAlister; Jacqui Mendez; Derek Ogg; Joe Patel; Philip Petteruti; Graeme R. Robb; Matthew B. Robers; Sakina Saif; Natalie Stratton; Dmitri I. Svergun; Wenxian Wang; David Whittaker; David Wilson; Yi Yao

Proteins of the bromodomain and extraterminal (BET) family, in particular bromodomain-containing protein 4 (BRD4), are of great interest as biological targets. BET proteins contain two separate bromodomains, and existing inhibitors bind to them monovalently. Here we describe the discovery and characterization of probe compound biBET, capable of engaging both bromodomains simultaneously in a bivalent, in cis binding mode. The evidence provided here was obtained in a variety of biophysical and cellular experiments. The bivalent binding results in very high cellular potency for BRD4 binding and pharmacological responses such as disruption of BRD4-mediator complex subunit 1 foci with an EC50 of 100 pM. These compounds will be of considerable utility as BET/BRD4 chemical probes. This work illustrates a novel concept in ligand design-simultaneous targeting of two separate domains with a drug-like small molecule-providing precedent for a potentially more effective paradigm for developing ligands for other multi-domain proteins.


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.


Molecular Cancer Therapeutics | 2016

AZD5153: a novel bivalent BET bromodomain inhibitor highly active against hematologic malignancies

Garrett W. Rhyasen; Maureen Hattersley; Yi Yao; Austin Dulak; Wenxian Wang; Philip Petteruti; Ian L. Dale; Scott Boiko; Tony Cheung; Jingwen Zhang; Shenghua Wen; Lillian Castriotta; Deborah Lawson; Mike Collins; Larry Bao; Miika Ahdesmaki; Graeme Walker; Greg O'Connor; Tammie C. Yeh; Alfred A. Rabow; Jonathan R. Dry; Corinne Reimer; Paul Lyne; Gordon B. Mills; Stephen Fawell; Michael J. Waring; Michael Zinda; Edwin Clark; Huawei Chen

The bromodomain and extraterminal (BET) protein BRD4 regulates gene expression via recruitment of transcriptional regulatory complexes to acetylated chromatin. Pharmacological targeting of BRD4 bromodomains by small molecule inhibitors has proven to be an effective means to disrupt aberrant transcriptional programs critical for tumor growth and/or survival. Herein, we report AZD5153, a potent, selective, and orally available BET/BRD4 bromodomain inhibitor possessing a bivalent binding mode. Unlike previously described monovalent inhibitors, AZD5153 ligates two bromodomains in BRD4 simultaneously. The enhanced avidity afforded through bivalent binding translates into increased cellular and antitumor activity in preclinical hematologic tumor models. In vivo administration of AZD5153 led to tumor stasis or regression in multiple xenograft models of acute myeloid leukemia, multiple myeloma, and diffuse large B-cell lymphoma. The relationship between AZD5153 exposure and efficacy suggests that prolonged BRD4 target coverage is a primary efficacy driver. AZD5153 treatment markedly affects transcriptional programs of MYC, E2F, and mTOR. Of note, mTOR pathway modulation is associated with cell line sensitivity to AZD5153. Transcriptional modulation of MYC and HEXIM1 was confirmed in AZD5153-treated human whole blood, thus supporting their use as clinical pharmacodynamic biomarkers. This study establishes AZD5153 as a highly potent, orally available BET/BRD4 inhibitor and provides a rationale for clinical development in hematologic malignancies. Mol Cancer Ther; 15(11); 2563–74. ©2016 AACR.


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.


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.


MedChemComm | 2012

Enantiomeric pairs reveal that key medicinal chemistry parameters vary more than simple physical property based models can explain

Andrew G. Leach; Elizabeth A. Pilling; Alfred A. Rabow; Simone Tomasi; Nabil Asaad; Niklaas J. Buurma; Andrew Ballard; Stefania Narduolo

An on-going trend in the medicinal chemistry literature is to link physical properties of a molecule that are achiral (such as lipophilicity) to biological or pharmaceutical properties (such as pharmacokinetics and toxicity) which depend upon interactions with chiral entities. In contrast, we have explored the variation in key pharmaceutical properties between enantiomeric pairs where only the three-dimensional arrangement of atoms changes. The analysis reveals that many properties are sensitive to changes in stereochemistry and quantifies this dependence. Any variation between enantiomers cannot be explained by achiral descriptors. The analysis highlights those properties where chiral lability or measurements on racemates might be misleading and permits deductions to be made from paired experiments in which two enantiomers are studied. The analysis suggests an inherent advantage to lead optimization in a chiral series where improvement into an attractive part of chemical space might be complemented by a selection between two enantiomers with different pharmacokinetic or safety profiles. Hence, in the early stages of a drug discovery program extra preference should be given to chiral series. Surprising observations are that permeability and efflux in cell based assays are not chirally dependent whereas in vivo volume of distribution does change between enantiomers.

Collaboration


Dive into the Alfred A. Rabow's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David G. Covell

Science Applications International Corporation

View shared research outputs
Researchain Logo
Decentralizing Knowledge