Network


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

Hotspot


Dive into the research topics where Graham Michael Wynne is active.

Publication


Featured researches published by Graham Michael Wynne.


Journal of Medicinal Chemistry | 2011

Discovery of 2-arylbenzoxazoles as upregulators of utrophin production for the treatment of Duchenne muscular dystrophy.

Chancellor; Kay E. Davies; O De Moor; Colin Richard Dorgan; Peter David Johnson; Adam Lambert; D Lawrence; Cristina Lecci; C Maillol; Penny Middleton; Gary Nugent; Severine Danielle Poignant; A Potter; Paul Damien Price; Richard J. Pye; Richard Storer; Jonathon M. Tinsley; R van Well; Richard Vickers; J Vile; Fraser Wilkes; Francis X. Wilson; Stephen Paul Wren; Graham Michael Wynne

A series of novel 2-arylbenzoxazoles that upregulate the production of utrophin in murine H2K cells, as assessed using a luciferase reporter linked assay, have been identified. This compound class appears to hold considerable promise as a potential treatment for Duchenne muscular dystrophy. Following the delineation of structure-activity relationships in the series, a number of potent upregulators were identified, and preliminary ADME evaluation is described. These studies have resulted in the identification of 1, a compound that has been progressed to clinical trials.


Human Molecular Genetics | 2015

Second-generation compound for the modulation of utrophin in the therapy of DMD

Simon Guiraud; Sarah Squire; Benjamin Edwards; H. Chen; D. Burns; Nandini Shah; Arran Babbs; Stephen G. Davies; Graham Michael Wynne; Angela J. Russell; David Elsey; Francis X. Wilson; Jon Tinsley; Kay E. Davies

Duchenne muscular dystrophy (DMD) is a lethal, X-linked muscle-wasting disease caused by lack of the cytoskeletal protein dystrophin. There is currently no cure for DMD although various promising approaches are progressing through human clinical trials. By pharmacologically modulating the expression of the dystrophin-related protein utrophin, we have previously demonstrated in dystrophin-deficient mdx studies, daily SMT C1100 treatment significantly reduced muscle degeneration leading to improved muscle function. This manuscript describes the significant disease modifying benefits associated with daily dosing of SMT022357, a second-generation compound in this drug series with improved physicochemical properties and a more robust metabolism profile. These studies in the mdx mouse demonstrate that oral administration of SMT022357 leads to increased utrophin expression in skeletal, respiratory and cardiac muscles. Significantly, utrophin expression is localized along the length of the muscle fibre, not just at the synapse, and is fibre-type independent, suggesting that drug treatment is modulating utrophin transcription in extra-synaptic myonuclei. This results in improved sarcolemmal stability and prevents dystrophic pathology through a significant reduction of regeneration, necrosis and fibrosis. All these improvements combine to protect the mdx muscle from contraction induced damage and enhance physiological function. This detailed evaluation of the SMT C1100 drug series strongly endorses the therapeutic potential of utrophin modulation as a disease modifying therapeutic strategy for all DMD patients irrespective of their dystrophin mutation.


Journal of Medicinal Chemistry | 2015

Stemistry: the control of stem cells in situ using chemistry.

Stephen G. Davies; Peter D. Kennewell; Angela J. Russell; Peter T. Seden; Robert Westwood; Graham Michael Wynne

A new paradigm for drug research has emerged, namely the deliberate search for molecules able to selectively affect the proliferation, differentiation, and migration of adult stem cells within the tissues in which they exist. Recently, there has been significant interest in medicinal chemistry toward the discovery and design of low molecular weight molecules that affect stem cells and thus have novel therapeutic activity. We believe that a successful agent from such a discover program would have profound effects on the treatment of many long-term degenerative disorders. Among these conditions are examples such as cardiovascular decay, neurological disorders including Alzheimers disease, and macular degeneration, all of which have significant unmet medical needs. This perspective will review evidence from the literature that indicates that discovery of such agents is achievable and represents a worthwhile pursuit for the skills of the medicinal chemist.


Bioorganic & Medicinal Chemistry Letters | 2011

Discovery and SAR of 2-arylbenzotriazoles and 2-arylindazoles as potential treatments for Duchenne muscular dystrophy.

Olivier De Moor; Colin Richard Dorgan; Peter David Johnson; Adam Lambert; Cristina Lecci; Carole Maillol; Gary Nugent; Severine Danielle Poignant; Paul Damien Price; Richard J. Pye; Richard Storer; Jonathon M. Tinsley; Richard Vickers; Renate van Well; Fraser Wilkes; Francis X. Wilson; Stephen Paul Wren; Graham Michael Wynne

Families of 2-arylbenzotriazoles and 2-arylindazoles that show positive effects in screens predictive of endogenous utrophin upregulation have been identified. Synthesis and structure-activity relationships are described leading to compounds with attractive in vitro profiles.


Bioorganic & Medicinal Chemistry | 2015

Ligand-based virtual screening identifies a family of selective cannabinoid receptor 2 agonists

Matteo Gianella-Borradori; Ivy Christou; Carole J. R. Bataille; Rebecca L. Cross; Graham Michael Wynne; David R. Greaves; Angela J. Russell

Graphical abstract


Bioorganic & Medicinal Chemistry | 2014

Structure-activity relationships and colorimetric properties of specific probes for the putative cancer biomarker human arylamine N-acetyltransferase 1.

James E. Egleton; Cyrille C. Thinnes; Peter T. Seden; Nicola Laurieri; Siu Po Lee; Kate S. Hadavizadeh; Angelina R. Measures; Alan M. Jones; Sam Thompson; Amy Varney; Graham Michael Wynne; Ali Ryan; Edith Sim; Angela J. Russell

A naphthoquinone inhibitor of human arylamine N-acetyltransferase 1 (hNAT1), a potential cancer biomarker and therapeutic target, has been reported which undergoes a distinctive concomitant color change from red to blue upon binding to the enzyme. Here we describe the use of in silico modeling alongside structure-activity relationship studies to advance the hit compound towards a potential probe to quantify hNAT1 levels in tissues. Derivatives with both a fifty-fold higher potency against hNAT1 and a two-fold greater absorption coefficient compared to the initial hit have been synthesized; these compounds retain specificity for hNAT1 and its murine homologue mNat2 over the isoenzyme hNAT2. A relationship between pKa, inhibitor potency and colorimetric properties has also been uncovered. The high potency of representative examples against hNAT1 in ZR-75-1 cell extracts also paves the way for the development of inhibitors with improved intrinsic sensitivity which could enable detection of hNAT1 in tissue samples and potentially act as tools for elucidating the unknown role hNAT1 plays in ER+ breast cancer; this could in turn lead to a therapeutic use for such inhibitors.


Bioorganic & Medicinal Chemistry | 2017

Thiazolidine derivatives as potent and selective inhibitors of the PIM kinase family

Carole J. R. Bataille; Méabh B. Brennan; Simon Byrne; Stephen G. Davies; Matthew J. Durbin; Oleg Fedorov; Kilian Huber; Alan M. Jones; Stefan Knapp; Gu Liu; Anna Nadali; Camilo E. Quevedo; Angela J. Russell; Roderick G. Walker; Robert Westwood; Graham Michael Wynne

The PIM family of serine/threonine kinases have become an attractive target for anti-cancer drug development, particularly for certain hematological malignancies. Here, we describe the discovery of a series of inhibitors of the PIM kinase family using a high throughput screening strategy. Through a combination of molecular modeling and optimization studies, the intrinsic potencies and molecular properties of this series of compounds was significantly improved. An excellent pan-PIM isoform inhibition profile was observed across the series, while optimized examples show good selectivity over other kinases. Two PIM-expressing leukemic cancer cell lines, MV4-11 and K562, were employed to evaluate the in vitro anti-proliferative effects of selected inhibitors. Encouraging activities were observed for many examples, with the best example (44) giving an IC50 of 0.75μM against the K562 cell line. These data provide a promising starting point for further development of this series as a new cancer therapy through PIM kinase inhibition.


PLOS Neglected Tropical Diseases | 2017

Dihydrobenz[e][1,4]oxazepin-2(3H)-ones, a new anthelmintic chemotype immobilising whipworm and reducing infectivity in vivo

Frederick A. Partridge; Emma A. Murphy; Nicky J. Willis; Carole J. R. Bataille; Ruth Forman; Narinder Heyer-Chauhan; Bruno Marinič; Daniel J. C. Sowood; Graham Michael Wynne; Kathryn J. Else; Angela J. Russell; David B. Sattelle

Trichuris trichiura is a human parasitic whipworm infecting around 500 million people globally, damaging the physical growth and educational performance of those infected. Current drug treatment options are limited and lack efficacy against the worm, preventing an eradication programme. It is therefore important to develop new treatments for trichuriasis. Using Trichuris muris, an established model for T. trichiura, we screened a library of 480 novel drug-like small molecules for compounds causing paralysis of the ex vivo adult parasite. We identified a class of dihydrobenz[e][1,4]oxazepin-2(3H)-one compounds with anthelmintic activity against T. muris. Further screening of structurally related compounds and resynthesis of the most potent molecules led to the identification of 20 active dihydrobenzoxazepinones, a class of molecule not previously implicated in nematode control. The most active immobilise adult T. muris with EC50 values around 25–50μM, comparable to the existing anthelmintic levamisole. The best compounds from this chemotype show low cytotoxicity against murine gut epithelial cells, demonstrating selectivity for the parasite. Developing a novel oral pharmaceutical treatment for a neglected disease and deploying it via mass drug administration is challenging. Interestingly, the dihydrobenzoxazepinone OX02983 reduces the ability of embryonated T. muris eggs to establish infection in the mouse host in vivo. Complementing the potential development of dihydrobenzoxazepinones as an oral anthelmintic, this supports an alternative strategy of developing a therapeutic that acts in the environment, perhaps via a spray, to interrupt the parasite lifecycle. Together these results show that the dihydrobenzoxazepinones are a new class of anthelmintic, active against both egg and adult stages of Trichuris parasites. They demonstrate encouraging selectivity for the parasite, and importantly show considerable scope for further optimisation to improve potency and pharmacokinetic properties with the aim of developing a clinical agent.


International Journal for Parasitology-Drugs and Drug Resistance | 2018

An automated high-throughput system for phenotypic screening of chemical libraries on C. elegans and parasitic nematodes.

Frederick A. Partridge; Anwen E. Brown; Steven D. Buckingham; Nicky J. Willis; Graham Michael Wynne; Ruth Forman; Kathryn J. Else; Alison A. Morrison; Jacqueline B. Matthews; Angela J. Russell; David A. Lomas; David B. Sattelle

Parasitic nematodes infect hundreds of millions of people and farmed livestock. Further, plant parasitic nematodes result in major crop damage. The pipeline of therapeutic compounds is limited and parasite resistance to the existing anthelmintic compounds is a global threat. We have developed an INVertebrate Automated Phenotyping Platform (INVAPP) for high-throughput, plate-based chemical screening, and an algorithm (Paragon) which allows screening for compounds that have an effect on motility and development of parasitic worms. We have validated its utility by determining the efficacy of a panel of known anthelmintics against model and parasitic nematodes: Caenorhabditis elegans, Haemonchus contortus, Teladorsagia circumcincta, and Trichuris muris. We then applied the system to screen the Pathogen Box chemical library in a blinded fashion and identified compounds already known to have anthelmintic or anti-parasitic activity, including tolfenpyrad, auranofin, and mebendazole; and 14 compounds previously undescribed as anthelmintics, including benzoxaborole and isoxazole chemotypes. This system offers an effective, high-throughput system for the discovery of novel anthelmintics.


Frontiers in Immunology | 2018

Activation of the immune-metabolic receptor GPR84 enhances inflammation and phagocytosis in macrophages

Carlota Recio; D Lucy; Purvis Gsd.; P Iveson; L Zeboudj; Asif J. Iqbal; Da Lin; Christopher A. O'Callaghan; L Davison; E Griesbach; Angela J. Russell; Graham Michael Wynne; L Dib; Claudia Monaco; David Robert Greaves

GPR84 is a member of the metabolic G protein-coupled receptor family, and its expression has been described predominantly in immune cells. GPR84 activation is involved in the inflammatory response, but the mechanisms by which it modulates inflammation have been incompletely described. In this study, we investigated GPR84 expression, activation, and function in macrophages to establish the role of the receptor during the inflammatory response. We observed that GPR84 expression in murine tissues is increased by endotoxemia, hyperglycemia, and hypercholesterolemia. Ex vivo studies revealed that GPR84 mRNA expression is increased by LPS and other pro-inflammatory molecules in different murine and human macrophage populations. Likewise, high glucose concentrations and the presence of oxidized LDL increased GPR84 expression in macrophages. Activation of the GPR84 receptor with a selective agonist, 6-(octylamino) pyrimidine-2,4(1H,3H)-dione (6-n-octylaminouracil, 6-OAU), enhanced the expression of phosphorylated Akt, p-ERK, and p65 nuclear translocation under inflammatory conditions and elevated the expression levels of the inflammatory mediators TNFα, IL-6, IL-12B, CCL2, CCL5, and CXCL1. In addition, GPR84 activation triggered increased bacterial adhesion and phagocytosis in macrophages. The enhanced inflammatory response mediated by 6-OAU was not observed in GPR84−/− cells nor in macrophages treated with a selective GPR84 antagonist. Collectively, our results reveal that GPR84 functions as an enhancer of inflammatory signaling in macrophages once inflammation is established. Therefore, molecules that antagonize the GPR84 receptor may be potential therapeutic tools in inflammatory and metabolic diseases.

Collaboration


Dive into the Graham Michael Wynne's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Richard Storer

University of Hertfordshire

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge