Philip Rawlins
AstraZeneca
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Philip Rawlins.
Nature Chemical Biology | 2016
Gizem Akçay; Matthew A. Belmonte; Brian Aquila; Claudio Chuaqui; Alexander Hird; Michelle L. Lamb; Philip Rawlins; Nancy Su; Sharon Tentarelli; Neil Grimster; Qibin Su
Targeted covalent inhibition of disease-associated proteins has become a powerful methodology in the field of drug discovery, leading to the approval of new therapeutics. Nevertheless, current approaches are often limited owing to their reliance on a cysteine residue to generate the covalent linkage. Here we used aryl boronic acid carbonyl warheads to covalently target a noncatalytic lysine side chain, and generated to our knowledge the first reversible covalent inhibitors for Mcl-1, a protein-protein interaction (PPI) target that has proven difficult to inhibit via traditional medicinal chemistry strategies. These covalent binders exhibited improved potency in comparison to noncovalent congeners, as demonstrated in biochemical and cell-based assays. We identified Lys234 as the residue involved in covalent modification, via point mutation. The covalent binders discovered in this study will serve as useful starting points for the development of Mcl-1 therapeutics and probes to interrogate Mcl-1-dependent biological phenomena.
Drug Discovery Today | 2016
J. Daniel Hothersall; Alastair J. H. Brown; Ian L. Dale; Philip Rawlins
Residence time describes the how long a ligand is bound to its target, and is attracting interest in drug discovery as a potential means of improving clinical efficacy by increasing target coverage. This concept, as originally applied to antagonists, is more complicated for G-protein-coupled receptor (GPCR) agonists because of the transiency of receptor responses (via desensitization and internalization). However, in some cases sustained GPCR agonist responses have been observed, with evidence consistent with a role for slow binding kinetics. We propose a model to explain our understanding of how residence time and rebinding might influence sustained signaling by internalized receptors. We also highlight the anticipated benefit for drug discovery of fully understanding and exploiting these phenomena to target desirable receptor response profiles selectively.
Journal of Medicinal Chemistry | 2017
William Mccoull; Roman D. Abrams; Erica Anderson; Kevin Blades; Peter Barton; Matthew R. Box; Jonathan Burgess; Kate Byth; Qing Cao; Claudio Chuaqui; Rodrigo J. Carbajo; Tony Cheung; Erin Code; Andrew D. Ferguson; Shaun Fillery; Nathan O. Fuller; Eric Gangl; Ning Gao; Matthew Grist; David Hargreaves; Martin R. Howard; Jun Hu; Paul D. Kemmitt; Jennifer E. Nelson; Nichole O’Connell; D. Bryan Prince; Piotr Raubo; Philip Rawlins; Graeme R. Robb; Junjie Shi
Inhibition of the protein-protein interaction between B-cell lymphoma 6 (BCL6) and corepressors has been implicated as a therapeutic target in diffuse large B-cell lymphoma (DLBCL) cancers and profiling of potent and selective BCL6 inhibitors are critical to test this hypothesis. We identified a pyrazolo[1,5-a]pyrimidine series of BCL6 binders from a fragment screen in parallel with a virtual screen. Using structure-based drug design, binding affinity was increased 100000-fold. This involved displacing crystallographic water, forming new ligand-protein interactions and a macrocyclization to favor the bioactive conformation of the ligands. Optimization for slow off-rate constant kinetics was conducted as well as improving selectivity against an off-target kinase, CK2. Potency in a cellular BCL6 assay was further optimized to afford highly selective probe molecules. Only weak antiproliferative effects were observed across a number of DLBCL lines and a multiple myeloma cell line without a clear relationship to BCL6 potency. As a result, we conclude that the BCL6 hypothesis in DLBCL cancer remains unproven.
ACS Medicinal Chemistry Letters | 2017
Jeffrey W. Johannes; Stephanie Bates; Carl Beigie; Matthew A. Belmonte; John Breen; Shenggen Cao; Paolo A. Centrella; Matthew A. Clark; John W. Cuozzo; Christoph E. Dumelin; Andrew D. Ferguson; Sevan Habeshian; David Hargreaves; Camil Joubran; Steven Kazmirski; Anthony D. Keefe; Michelle L. Lamb; Haiye Lan; Yunxia Li; Hao Ma; Scott Mlynarski; Martin J. Packer; Philip Rawlins; Daniel W. Robbins; Haidong Shen; Eric A. Sigel; Holly H. Soutter; Nancy Su; Dawn M. Troast; Haiyun Wang
Mcl-1 is a pro-apoptotic BH3 protein family member similar to Bcl-2 and Bcl-xL. Overexpression of Mcl-1 is often seen in various tumors and allows cancer cells to evade apoptosis. Here we report the discovery and optimization of a series of non-natural peptide Mcl-1 inhibitors. Screening of DNA-encoded libraries resulted in hit compound 1, a 1.5 μM Mcl-1 inhibitor. A subsequent crystal structure demonstrated that compound 1 bound to Mcl-1 in a β-turn conformation, such that the two ends of the peptide were close together. This proximity allowed for the linking of the two ends of the peptide to form a macrocycle. Macrocyclization resulted in an approximately 10-fold improvement in binding potency. Further exploration of a key hydrophobic interaction with Mcl-1 protein and also with the moiety that engages Arg256 led to additional potency improvements. The use of protein-ligand crystal structures and binding kinetics contributed to the design and understanding of the potency gains. Optimized compound 26 is a <3 nM Mcl-1 inhibitor, while inhibiting Bcl-2 at only 5 μM and Bcl-xL at >99 μM, and induces cleaved caspase-3 in MV4-11 cells with an IC50 of 3 μM after 6 h.
Molecular Pharmacology | 2016
J. D. Hothersall; Dong Guo; S. Sarda; R. J. Sheppard; Huawei Chen; W. Keur; Michael J. Waring; Adriaan P. IJzerman; Stephen J. Hill; Ian L. Dale; Philip Rawlins
The duration of action of adenosine A2A receptor (A2A) agonists is critical for their clinical efficacy, and we sought to better understand how this can be optimized. The in vitro temporal response profiles of a panel of A2A agonists were studied using cAMP assays in recombinantly (CHO) and endogenously (SH-SY5Y) expressing cells. Some agonists (e.g., 3cd; UK-432,097) but not others (e.g., 3ac; CGS-21680) demonstrated sustained wash-resistant agonism, where residual receptor activation continued after washout. The ability of an antagonist to reverse pre-established agonist responses was used as a surrogate read-out for agonist dissociation kinetics, and together with radioligand binding studies suggested a role for slow off-rate in driving sustained effects. One compound, 3ch, showed particularly marked sustained effects, with a reversal t1/2 > 6 hours and close to maximal effects that remained for at least 5 hours after washing. Based on the structure-activity relationship of these compounds, we suggest that lipophilic N6 and bulky C2 substituents can promote stable and long-lived binding events leading to sustained agonist responses, although a high compound logD is not necessary. This provides new insight into the binding interactions of these ligands and we anticipate that this information could facilitate the rational design of novel long-acting A2A agonists with improved clinical efficacy.
European Journal of Pharmacology | 2015
J. Daniel Hothersall; Charlotte E. Bussey; Alastair J. H. Brown; James S. Scott; Ian L. Dale; Philip Rawlins
G protein-coupled receptor 119 (GPR119) is involved in regulating metabolic homoeostasis, with GPR119 agonists targeted for the treatment of type-2 diabetes and obesity. Using the endogenous agonist oleoylethanolamide and a number of small molecule synthetic agonists we have investigated the temporal dynamics of receptor signalling. Using both a dynamic luminescence biosensor-based assay and an endpoint cAMP accumulation assay we show that agonist-driven desensitization is not a major regulatory mechanism for GPR119 despite robust activation responses, regardless of the agonist used. Temporal analysis of the cAMP responses demonstrated sustained signalling resistant to washout for some, but not all of the agonists tested. Further analysis indicated that the sustained effects of one synthetic agonist AR-231,453 were consistent with a role for slow dissociation kinetics. In contrast, the sustained responses to MBX-2982 and AZ1 appeared to involve membrane deposition. We also detect wash-resistant responses to AR-231,453 at the level of physiologically relevant responses in an endogenous expression system (GLP-1 secretion in GLUTag cells). In conclusion, our findings indicate that in a recombinant expression system GPR119 activation is sustained, with little evidence of pronounced receptor desensitization, and for some ligands persistent agonist responses continue despite removal of excess agonist. This provides novel understanding of the temporal responses profiles of potential drug candidates targetting GPR119, and highlights the importance of carefully examining the the mechanisms through which GPCRs generate sustained responses.
ACS Chemical Biology | 2018
William Mccoull; Tony Cheung; Erica Anderson; Peter Barton; Jonathan Burgess; Kate Byth; Qing Cao; M. Paola Castaldi; Huawei Chen; Elisabetta Chiarparin; Rodrigo J. Carbajo; Erin Code; Suzanna Cowan; Paul R.J. Davey; Andrew D. Ferguson; Shaun Fillery; Nathan O. Fuller; Ning Gao; David Hargreaves; Martin R. Howard; Jun Hu; Aarti Kawatkar; Paul D. Kemmitt; Elisabetta Leo; Daniel M. Molina; Nichole O’Connell; Philip Petteruti; Timothy Rasmusson; Piotr Raubo; Philip Rawlins
B-cell lymphoma 6 (BCL6) inhibition is a promising mechanism for treating hematological cancers but high quality chemical probes are necessary to evaluate its therapeutic potential. Here we report potent BCL6 inhibitors that demonstrate cellular target engagement and exhibit exquisite selectivity for BCL6 based on mass spectrometry analyses following chemical proteomic pull down. Importantly, a proteolysis-targeting chimera (PROTAC) was also developed and shown to significantly degrade BCL6 in a number of diffuse large B-cell lymphoma (DLBCL) cell lines, but neither BCL6 inhibition nor degradation selectively induced marked phenotypic response. To investigate, we monitored PROTAC directed BCL6 degradation in DLBCL OCI-Ly1 cells by immunofluorescence and discovered a residual BCL6 population. Analysis of subcellular fractions also showed incomplete BCL6 degradation in all fractions despite having measurable PROTAC concentrations, together providing a rationale for the weak antiproliferative response seen with both BCL6 inhibitor and degrader. In summary, we have developed potent and selective BCL6 inhibitors and a BCL6 PROTAC that effectively degraded BCL6, but both modalities failed to induce a significant phenotypic response in DLBCL despite achieving cellular concentrations.
Journal of Biomolecular Screening | 2017
Göran Dahl; Stephan Steigele; Per Hillertz; Anna Tigerström; Anders Egnéus; Alexander Mehrle; Martin Ginkel; Fredrik Edfeldt; Geoff Holdgate; Nichole O’Connell; Bernd Kappler; Annette Brodte; Philip Rawlins; Gareth Davies; Eva-Lotta Westberg; Rutger H. A. Folmer; Stephan Heyse
Surface plasmon resonance (SPR) is a powerful method for obtaining detailed molecular interaction parameters. Modern instrumentation with its increased throughput has enabled routine screening by SPR in hit-to-lead and lead optimization programs, and SPR has become a mainstream drug discovery technology. However, the processing and reporting of SPR data in drug discovery are typically performed manually, which is both time-consuming and tedious. Here, we present the workflow concept, design and experiences with a software module relying on a single, browser-based software platform for the processing, analysis, and reporting of SPR data. The efficiency of this concept lies in the immediate availability of end results: data are processed and analyzed upon loading the raw data file, allowing the user to immediately quality control the results. Once completed, the user can automatically report those results to data repositories for corporate access and quickly generate printed reports or documents. The software module has resulted in a very efficient and effective workflow through saved time and improved quality control. We discuss these benefits and show how this process defines a new benchmark in the drug discovery industry for the handling, interpretation, visualization, and sharing of SPR data.
ACS Medicinal Chemistry Letters | 2017
Jeffrey W. Johannes; Stephanie Bates; Carl Beigie; Matthew A. Belmonte; John Breen; Shenggen Cao; Paolo A. Centrella; Matthew A. Clark; John W. Cuozzo; Christoph E. Dumelin; Andrew D. Ferguson; Sevan Habeshian; David Hargreaves; Camil Joubran; Steven Kazmirski; Anthony D. Keefe; Michelle L. Lamb; Haiye Lan; Yunxia Li; Hao Ma; Scott Mlynarski; Martin J. Packer; Philip Rawlins; Daniel W. Robbins; Haidong Shen; Eric A. Sigel; Holly H. Soutter; Nancy Su; Dawn M. Troast; Haiyun Wang
[This corrects the article DOI: 10.1021/acsmedchemlett.6b00464.].
Molecular Cancer Therapeutics | 2015
Qibin Su; Gizem Akçay; Neil Grimster; Matthew A. Belmonte; Philip Rawlins; Michelle Lamb; Alexander Hird; Brian Aquila
Myeloid cell leukemia 1 (Mcl-1), a potent anti-apoptotic protein of the BCL2 family, has been studied as a key resistance factor in human cancers. Restoring apoptotic signals by inactivating Mcl-1 protein interactions with small molecule inhibitors has been intensively pursued as targets for cancer therapeutics. We herein describe our effort towards the structure-based design, synthesis and evaluation of first potent, covalent binders of Mcl-1. The resulting inhibitors specifically modified a non-catalytic lysine residue with high level of Mcl-1 potency in biochemical and cell based assays. Our covalent inhibitors could provide potent probes to interrogate Mcl-1 dependent cancer biology. Citation Format: Qibin Su, Gizem Akcay, Neil Grimster, Matthew Belmonte, Philip Rawlins, Michelle Lamb, Alexander Hird, Brian Aquila. Inhibition of Mcl-1 through covalent modification of a non-catalytic lysine side chain. [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 C40.