Network


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

Hotspot


Dive into the research topics where Thomas M. McGuire is active.

Publication


Featured researches published by Thomas M. McGuire.


Journal of Medicinal Chemistry | 2013

Discovery of 4-{4-[(3R)-3-Methylmorpholin-4-yl]-6-[1-(methylsulfonyl)cyclopropyl]pyrimidin-2-yl}-1H-indole (AZ20): a potent and selective inhibitor of ATR protein kinase with monotherapy in vivo antitumor activity.

Kevin Michael Foote; Kevin Blades; Anna Cronin; Shaun Fillery; Sylvie S. Guichard; Lorraine Hassall; Ian Hickson; Xavier Jacq; Philip J. Jewsbury; Thomas M. McGuire; J. Willem M. Nissink; Rajesh Odedra; Ken Page; Paula Perkins; Abid Suleman; Kin Yip Tam; Pia Thommes; Rebecca Broadhurst; Christine Wood

ATR is an attractive new anticancer drug target whose inhibitors have potential as chemo- or radiation sensitizers or as monotherapy in tumors addicted to particular DNA-repair pathways. We describe the discovery and synthesis of a series of sulfonylmorpholinopyrimidines that show potent and selective ATR inhibition. Optimization from a high quality screening hit within tight SAR space led to compound 6 (AZ20) which inhibits ATR immunoprecipitated from HeLa nuclear extracts with an IC50 of 5 nM and ATR mediated phosphorylation of Chk1 in HT29 colorectal adenocarcinoma tumor cells with an IC50 of 50 nM. Compound 6 potently inhibits the growth of LoVo colorectal adenocarcinoma tumor cells in vitro and has high free exposure in mouse following moderate oral doses. At well tolerated doses 6 leads to significant growth inhibition of LoVo xenografts grown in nude mice. Compound 6 is a useful compound to explore ATR pharmacology in vivo.


Journal of the American Chemical Society | 2016

KOtBu: A Privileged Reagent for Electron Transfer Reactions?

Joshua P. Barham; Graeme Coulthard; Katie J. Emery; Eswararao Doni; Florimond Cumine; Giuseppe Nocera; Matthew P. John; L.E.A. Berlouis; Thomas M. McGuire; Tell Tuttle; John A. Murphy

Many recent studies have used KOtBu in organic reactions that involve single electron transfer; in the literature, the electron transfer is proposed to occur either directly from the metal alkoxide or indirectly, following reaction of the alkoxide with a solvent or additive. These reaction classes include coupling reactions of halobenzenes and arenes, reductive cleavages of dithianes, and SRN1 reactions. Direct electron transfer would imply that alkali metal alkoxides are willing partners in these electron transfer reactions, but the literature reports provide little or no experimental evidence for this. This paper examines each of these classes of reaction in turn, and contests the roles proposed for KOtBu; instead, it provides new mechanistic information that in each case supports the in situ formation of organic electron donors. We go on to show that direct electron transfer from KOtBu can however occur in appropriate cases, where the electron acceptor has a reduction potential near the oxidation potential of KOtBu, and the example that we use is CBr4. In this case, computational results support electrochemical data in backing a direct electron transfer reaction.


Journal of Medicinal Chemistry | 2012

(1R,2R)-N-(1-cyanocyclopropyl)-2-(6-methoxy-1,3,4,5-tetrahydropyrido[4,3-b]indole-2-carbonyl)cyclohexanecarboxamide (AZD4996): a potent and highly selective cathepsin K inhibitor for the treatment of osteoarthritis.

Alexander G. Dossetter; Howard Beeley; Jonathan Bowyer; Calum R. Cook; James J. Crawford; Jonathan E. Finlayson; Nicola Murdoch Heron; Christine Heyes; Adrian J. Highton; Julian A. Hudson; Anja Jestel; Peter W. Kenny; Stephan Krapp; Scott Martin; Philip A. MacFaul; Thomas M. McGuire; Pablo Morentin Gutierrez; Andrew D. Morley; Jeffrey James Morris; Ken Page; Lyn Rosenbrier Ribeiro; Helen Sawney; Stefan Steinbacher; Caroline L. Smith; Madeleine Vickers

Directed screening of nitrile compounds revealed 3 as a highly potent cathepsin K inhibitor but with cathepsin S activity and very poor stability to microsomes. Synthesis of compounds with reduced molecular complexity, such as 7, revealed key SAR and demonstrated that baseline physical properties and in vitro stability were in fact excellent for this series. The tricycle carboline P3 unit was discovered by hypothesis-based design using existing structural information. Optimization using small substituents, knowledge from matched molecular pairs, and control of lipophilicity yielded compounds very close to the desired profile, of which 34 (AZD4996) was selected on the basis of pharmacokinetic profile.


Cancer Research | 2013

Abstract 2348: Discovery of AZD6738, a potent and selective inhibitor with the potential to test the clinical efficacy of ATR kinase inhibition in cancer patients.

Clifford David Jones; Kevin Blades; Kevin Michael Foote; Sylvie Guichard; Philip J. Jewsbury; Thomas M. McGuire; Johannes Wilhelmus Maria Nissink; Rajesh Odedra; Kin Yip Tam; Pia Thommes; Paul Turner; Gary Wilkinson; Christine Wood; James Yates

AZD6738 is a potent and selective inhibitor of ataxia telangiectasia and rad3 related (ATR) kinase with the potential to be used in the clinic. We report the discovery of a morpholino-pyrimidine series and its subsequent optimisation to the pre-clinical candidate AZD6738 which has a good overall balance of potency, selectivity, pharmacokinetic and biopharmaceutical properties suitable for oral dosing. ATR is a serine/threonine protein kinase involved in DNA damage response signalling caused by DNA replication associated stress. Activation of ATR at stalled replication forks leads to suppression of replication fork origin firing, promotes repair and S/G2-cell cycle checkpoints to prevent premature mitosis and maintain genomic integrity. Failure to resolve damage leads to genomic instability and if sufficiently high, cell death. Stalled replication forks may collapse leading to formation of DNA double stranded breaks and activation of the ataxia telangiectasia mutated (ATM) kinase. ATM works in conjunction with ATR to efficiently resolve replication associated DNA damage creating a co-dependency with loss of one leading to a greater reliance on the other to maintain genomic stability. ATM is frequently inactivated across B-cell malignancies, head and neck, breast and lung cancers through chromosomal deletion, promoter hypermethylation or mutation. ATM-deficient tumours are hypothesised to be more reliant on ATR for survival and specific inhibition of ATR may lead enhanced anti-tumour activity while minimizing normal tissue toxicity. AZD6378 has the potential to test the clinical efficacy of ATR inhibition in cancer patients as monotherapy against ATM-deficient tumours or in combination with chemo or radiotherapy. Citation Format: Clifford D. Jones, Kevin Blades, Kevin M. Foote, Sylvie M. Guichard, Philip J. Jewsbury, Thomas McGuire, Johannes W. Nissink, Rajesh Odedra, Kin Tam, Pia Thommes, Paul Turner, Gary Wilkinson, Christine Wood, James W. Yates. Discovery of AZD6738, a potent and selective inhibitor with the potential to test the clinical efficacy of ATR kinase inhibition in cancer patients. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 2348. doi:10.1158/1538-7445.AM2013-2348


ACS Medicinal Chemistry Letters | 2015

Pyrimidinone Nicotinamide Mimetics as Selective Tankyrase and Wnt Pathway Inhibitors Suitable for in Vivo Pharmacology

Jeffrey W. Johannes; Lynsie Almeida; Bernard Barlaam; P. Ann Boriack-Sjodin; Robert Casella; Rosemary A. Croft; Allan Dishington; Lakshmaiah Gingipalli; Chungang Gu; Janet Hawkins; Jane L. Holmes; Tina Howard; Jian Huang; Stephanos Ioannidis; Steven Kazmirski; Michelle L. Lamb; Thomas M. McGuire; Jane E. Moore; Derek Ogg; Anil Patel; Kurt Gordon Pike; Timothy Pontz; Graeme R. Robb; Nancy Su; Haiyun Wang; Xiaoyun Wu; Hai-Jun Zhang; Yue Zhang; Xiaolan Zheng; Tao Wang

The canonical Wnt pathway plays an important role in embryonic development, adult tissue homeostasis, and cancer. Germline mutations of several Wnt pathway components, such as Axin, APC, and ß-catenin, can lead to oncogenesis. Inhibition of the poly(ADP-ribose) polymerase (PARP) catalytic domain of the tankyrases (TNKS1 and TNKS2) is known to inhibit the Wnt pathway via increased stabilization of Axin. In order to explore the consequences of tankyrase and Wnt pathway inhibition in preclinical models of cancer and its impact on normal tissue, we sought a small molecule inhibitor of TNKS1/2 with suitable physicochemical properties and pharmacokinetics for hypothesis testing in vivo. Starting from a 2-phenyl quinazolinone hit (compound 1), we discovered the pyrrolopyrimidinone compound 25 (AZ6102), which is a potent TNKS1/2 inhibitor that has 100-fold selectivity against other PARP family enzymes and shows 5 nM Wnt pathway inhibition in DLD-1 cells. Moreover, compound 25 can be formulated well in a clinically relevant intravenous solution at 20 mg/mL, has demonstrated good pharmacokinetics in preclinical species, and shows low Caco2 efflux to avoid possible tumor resistance mechanisms.


Journal of Medicinal Chemistry | 2016

Discovery of Novel 3-Quinoline Carboxamides as Potent, Selective and Orally Bioavailable Inhibitors of Ataxia Telangiectasia Mutated (Atm) Kinase.

Sébastien L. Degorce; Bernard Christophe Barlaam; Elaine Cadogan; Allan Dishington; Richard Ducray; Steven C. Glossop; Lorraine Hassall; Franck Lach; Alan Lau; Thomas M. McGuire; Thorsten Nowak; Gilles Ouvry; Kurt Gordon Pike; Andrew G. Thomason

A novel series of 3-quinoline carboxamides has been discovered and optimized as selective inhibitors of the ataxia telangiectasia mutated (ATM) kinase. From a modestly potent HTS hit (4), we identified molecules such as 6-[6-(methoxymethyl)-3-pyridinyl]-4-{[(1R)-1-(tetrahydro-2H-pyran-4-yl)ethyl]amino}-3-quinolinecarboxamide (72) and 7-fluoro-6-[6-(methoxymethyl)pyridin-3-yl]-4-{[(1S)-1-(1-methyl-1H-pyrazol-3-yl)ethyl]amino}quinoline-3-carboxamide (74) as potent and highly selective ATM inhibitors with overall ADME properties suitable for oral administration. 72 and 74 constitute excellent oral tools to probe ATM inhibition in vivo. Efficacy in combination with the DSB-inducing agent irinotecan was observed in a disease relevant model.


Journal of Organic Chemistry | 2015

Synthesis of Azabicycles via Cascade Aza-Prins Reactions: Accessing the Indolizidine and Quinolizidine Cores

Freda K. I. Chio; Sebastien J. J. Guesne; Lorraine Hassall; Thomas M. McGuire; Adrian P. Dobbs

The first detailed studies of intramolecular aza-Prins and aza-silyl-Prins reactions, starting from acyclic materials, are reported. The methods allow rapid and flexible access toward an array of [6,5] and [6,6] aza-bicycles, which form the core skeletons of various alkaloids. On the basis of our findings on the aza-Prins and aza-silyl-Prins cyclizations, herein we present simple protocols for the intramolecular preparation of the azabicyclic cores of the indolizidines and quinolizidines using a one-pot cascade process of N-acyliminium ion formation followed by aza-Prins cyclization and either elimination or carbocation trapping. It is possible to introduce a range of different substituents into the heterocycles through a judicial choice of Lewis acid and solvent(s), with halo-, phenyl-, and amido-substituted azabicyclic products all being accessed through these highly diastereoselective processes.


Journal of Medicinal Chemistry | 2012

Pharmacokinetic benefits of 3,4-dimethoxy substitution of a phenyl ring and design of isosteres yielding orally available cathepsin K inhibitors.

James J. Crawford; Peter W. Kenny; Jonathan Bowyer; Calum R. Cook; Jonathan E. Finlayson; Christine Heyes; Adrian J. Highton; Julian A. Hudson; Anja Jestel; Stephan Krapp; Scott Martin; Philip A. MacFaul; Benjamin P. McDermott; Thomas M. McGuire; Andrew D. Morley; Jeffrey James Morris; Ken Page; Lyn Rosenbrier Ribeiro; Helen Sawney; Stefan Steinbacher; Caroline L. Smith; Alexander G. Dossetter

Rational structure-based design has yielded highly potent inhibitors of cathepsin K (Cat K) with excellent physical properties, selectivity profiles, and pharmacokinetics. Compounds with a 3,4-(CH₃O)₂Ph motif, such as 31, were found to have excellent metabolic stability and absorption profiles. Through metabolite identification studies, a reactive metabolite risk was identified with this motif. Subsequent structure-based design of isoteres culminated in the discovery of an optimized and balanced inhibitor (indazole, 38).


Journal of Medicinal Chemistry | 2018

The Identification of Potent, Selective, and Orally Available Inhibitors of Ataxia Telangiectasia Mutated (ATM) Kinase: The Discovery of AZD0156 (8-{6-[3-(Dimethylamino)propoxy]pyridin-3-yl}-3-methyl-1-(tetrahydro-2H-pyran-4-yl)-1,3-dihydro-2H-imidazo[4,5-c]quinolin-2-one)

Kurt Gordon Pike; Bernard Barlaam; Elaine Cadogan; Andrew Campbell; Yingxue Chen; Nicola Colclough; Nichola L. Davies; Camila de-Almeida; Sébastien L. Degorce; Myriam Didelot; Allan Dishington; Richard Ducray; Stephen T. Durant; Lorraine Hassall; Jane L. Holmes; Gareth Hughes; Philip A. MacFaul; Keith Raymond Mulholland; Thomas M. McGuire; Gilles Ouvry; Martin Pass; Graeme R. Robb; Natalie Stratton; Zhenhua Wang; Joanne Wilson; Baochang Zhai; Kang Zhao; Nidal Al-Huniti

ATM inhibitors, such as 7, have demonstrated the antitumor potential of ATM inhibition when combined with DNA double-strand break-inducing agents in mouse xenograft models. However, the properties of 7 result in a relatively high predicted clinically efficacious dose. In an attempt to minimize attrition during clinical development, we sought to identify ATM inhibitors with a low predicted clinical dose (<50 mg) and focused on strategies to increase both ATM potency and predicted human pharmacokinetic half-life (predominantly through the increase of volume of distribution). These efforts resulted in the discovery of 64 (AZD0156), an exceptionally potent and selective inhibitor of ATM based on an imidazo[4,5- c]quinolin-2-one core. 64 has good preclinical phamacokinetics, a low predicted clinical dose, and a high maximum absorbable dose. 64 has been shown to potentiate the efficacy of the approved drugs irinotecan and olaparib in disease relevant mouse models and is currently undergoing clinical evaluation with these agents.


Bioorganic & Medicinal Chemistry Letters | 2012

Isosteric replacements for benzothiazoles and optimisation to potent Cathepsin K inhibitors free from hERG channel inhibition

Alexander G. Dossetter; Jonathan Bowyer; Calum R. Cook; James J. Crawford; Jonathan E. Finlayson; Nicola Murdoch Heron; Christine Heyes; Adrian J. Highton; Julian A. Hudson; Anja Jestel; Stephan Krapp; Philip A. MacFaul; Thomas M. McGuire; Andrew D. Morley; Jeffrey James Morris; Ken Page; Lyn Rosenbrier Ribeiro; Helen Sawney; Stefan Steinbacher; Caroline Smith

The discovery of nitrile compound 4, a potent inhibitor of Cathepsin K (Cat K) with good bioavailability in dog is described. The compound was used to demonstrate target engagement and inhibition of Cat K in an in vivo dog PD model. The margin to hERG ion channel inhibition was deemed too low for a clinical candidate and an optimisation program to find isosteres or substitutions on benzothiazole group led to the discovery of 20, 24 and 27; all three free from hERG inhibition.

Collaboration


Dive into the Thomas M. McGuire'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

John A. Murphy

University of Strathclyde

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge