Network


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

Hotspot


Dive into the research topics where Martin Pass is active.

Publication


Featured researches published by Martin Pass.


Cancer Research | 2010

AZD8055 Is a Potent, Selective, and Orally Bioavailable ATP-Competitive Mammalian Target of Rapamycin Kinase Inhibitor with In vitro and In vivo Antitumor Activity

Christine M. Chresta; Barry R. Davies; Ian Hickson; Tom Harding; Sabina Cosulich; Susan E. Critchlow; John Vincent; Rebecca Ellston; Darren Jones; Patrizia Sini; Dominic James; Zoe Howard; Phillippa Dudley; Gareth Hughes; Lisa L. Smith; Sharon Maguire; Marc Geoffery Hummersone; Karine Malagu; Keith Menear; Richard Jenkins; Matt Jacobsen; Graeme Cameron Murray Smith; Sylvie Guichard; Martin Pass

The mammalian target of rapamycin (mTOR) kinase forms two multiprotein complexes, mTORC1 and mTORC2, which regulate cell growth, cell survival, and autophagy. Allosteric inhibitors of mTORC1, such as rapamycin, have been extensively used to study tumor cell growth, proliferation, and autophagy but have shown only limited clinical utility. Here, we describe AZD8055, a novel ATP-competitive inhibitor of mTOR kinase activity, with an IC50 of 0.8 nmol/L. AZD8055 showed excellent selectivity (approximately 1,000-fold) against all class I phosphatidylinositol 3-kinase (PI3K) isoforms and other members of the PI3K-like kinase family. Furthermore, there was no significant activity against a panel of 260 kinases at concentrations up to 10 micromol/L. AZD8055 inhibits the phosphorylation of mTORC1 substrates p70S6K and 4E-BP1 as well as phosphorylation of the mTORC2 substrate AKT and downstream proteins. The rapamycin-resistant T37/46 phosphorylation sites on 4E-BP1 were fully inhibited by AZD8055, resulting in significant inhibition of cap-dependent translation. In vitro, AZD8055 potently inhibits proliferation and induces autophagy in H838 and A549 cells. In vivo, AZD8055 induces a dose-dependent pharmacodynamic effect on phosphorylated S6 and phosphorylated AKT at plasma concentrations leading to tumor growth inhibition. Notably, AZD8055 results in significant growth inhibition and/or regression in xenografts, representing a broad range of human tumor types. AZD8055 is currently in phase I clinical trials.


Molecular Cancer Therapeutics | 2012

Preclinical Pharmacology of AZD5363, an Inhibitor of AKT: Pharmacodynamics, Antitumor Activity, and Correlation of Monotherapy Activity with Genetic Background

Barry R. Davies; Hannah Greenwood; Philippa Dudley; Claire Crafter; De-Hua Yu; Jingchuan Zhang; Jing Li; Beirong Gao; Qunsheng Ji; Juliana Maynard; Sally-Ann Ricketts; Darren Cross; Sabina Cosulich; Christine M. Chresta; Ken Page; James Yates; Clare Lane; Rebecca Watson; Richard William Arthur Luke; Donald J. Ogilvie; Martin Pass

AKT is a key node in the most frequently deregulated signaling network in human cancer. AZD5363, a novel pyrrolopyrimidine-derived compound, inhibited all AKT isoforms with a potency of 10 nmol/L or less and inhibited phosphorylation of AKT substrates in cells with a potency of approximately 0.3 to 0.8 μmol/L. AZD5363 monotherapy inhibited the proliferation of 41 of 182 solid and hematologic tumor cell lines with a potency of 3 μmol/L or less. Cell lines derived from breast cancers showed the highest frequency of sensitivity. There was a significant relationship between the presence of PIK3CA and/or PTEN mutations and sensitivity to AZD5363 and between RAS mutations and resistance. Oral dosing of AZD5363 to nude mice caused dose- and time-dependent reduction of PRAS40, GSK3β, and S6 phosphorylation in BT474c xenografts (PRAS40 phosphorylation EC50 ∼ 0.1 μmol/L total plasma exposure), reversible increases in blood glucose concentrations, and dose-dependent decreases in 2[18F]fluoro-2-deoxy-d-glucose (18F-FDG) uptake in U87-MG xenografts. Chronic oral dosing of AZD5363 caused dose-dependent growth inhibition of xenografts derived from various tumor types, including HER2+ breast cancer models that are resistant to trastuzumab. AZD5363 also significantly enhanced the antitumor activity of docetaxel, lapatinib, and trastuzumab in breast cancer xenografts. It is concluded that AZD5363 is a potent inhibitor of AKT with pharmacodynamic activity in vivo, has potential to treat a range of solid and hematologic tumors as monotherapy or a combinatorial agent, and has potential for personalized medicine based on the genetic status of PIK3CA, PTEN, and RAS. AZD5363 is currently in phase I clinical trials. Mol Cancer Ther; 11(4); 873–87. ©2012 AACR.


Bioorganic & Medicinal Chemistry Letters | 2013

Optimization of potent and selective dual mTORC1 and mTORC2 inhibitors: The discovery of AZD8055 and AZD2014

Kurt Gordon Pike; Karine Malagu; Marc Geoffery Hummersone; Keith Menear; Heather Mary Ellen Duggan; Sylvie Gomez; Niall Morrison Barr Martin; Linette Ruston; Sarah L. Pass; Martin Pass

The optimization of a potent and highly selective series of dual mTORC1 and mTORC2 inhibitors is described. An initial focus on improving cellular potency whilst maintaining or improving other key parameters, such as aqueous solubility and margins over hERG IC(50), led to the discovery of the clinical candidate AZD8055 (14). Further optimization, particularly aimed at reducing the rate of metabolism in human hepatocyte incubations, resulted in the discovery of the clinical candidate AZD2014 (21).


Toxicologic Pathology | 2011

Induction of Heart Valve Lesions by Small-Molecule ALK5 Inhibitors

Mark J. Anderton; Howard R. Mellor; Alex R. Bell; Claire Sadler; Martin Pass; Steve Powell; Samantha J. Steele; Ruth A. Roberts; Annabelle Heier

Aberrant signaling by transforming growth factor-β (TGF-β) and its type I (ALK5) receptor has been implicated in a number of human diseases and this pathway is considered a potential target for therapeutic intervention. Transforming growth factor-β signaling via ALK5 plays a critical role during heart development, but the role of ALK5 in the adult heart is poorly understood. In the current study, the preclinical toxicology of ALK5 inhibitors from two different chemistry scaffolds was explored. Ten-week-old female Han Wistar rats received test compounds by the oral route for three to seven days. Both compounds induced histopathologic heart valve lesions characterized by hemorrhage, inflammation, degeneration, and proliferation of valvular interstitial cells. The pathology was observed in all animals, at all doses tested, and occurred in all four heart valves. Immunohistochemical analysis of ALK5 in rat hearts revealed expression in the valves, but not in the myocardium. Compared to control animals, protein levels of ALK5 were unchanged in the heart valves of treated animals. We also observed a physeal dysplasia in the femoro-tibial joint of rats treated with ALK5 inhibitors, a finding consistent with a pharmacological effect described previously with ALK5 inhibitors. Overall, these findings suggest that TGF-β signaling via ALK5 plays a critical role in maintaining heart valve integrity.


Bioorganic & Medicinal Chemistry Letters | 2009

The discovery and optimisation of pyrido[2,3-d]pyrimidine-2,4-diamines as potent and selective inhibitors of mTOR kinase.

Karine Malagu; Heather Mary Ellen Duggan; Keith Menear; Marc Geoffery Hummersone; Sylvie Gomez; Christine Bailey; Peter D. Edwards; Jan Drzewiecki; Frederic Leroux; Mar Jimenez Quesada; Gesine Johanna Hermann; Stephanie Maine; Carrie-Anne Molyneaux; Armelle Le Gall; James R. Pullen; Ian D. Hickson; Lisa L. Smith; Sharon Maguire; Niall Morrison Barr Martin; Graeme Smith; Martin Pass

We describe a novel series of potent inhibitors of the kinase activity of mTOR. The compounds display good selectivity relative to other PI3K-related kinase family members and, in cellular assays, inhibit both mTORC1 and mTORC2 complexes and exhibit good antiproliferative activity.


Journal of Medicinal Chemistry | 2013

Discovery of 4-Amino-N-[(1S)-1-(4-chlorophenyl)-3-hydroxypropyl]-1-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)piperidine-4-carboxamide (AZD5363), an Orally Bioavailable, Potent Inhibitor of Akt Kinases.

Matt Addie; Peter Ballard; David Buttar; Claire Crafter; Gordon S. Currie; Barry R. Davies; J.E. Debreczeni; Hannah Dry; Philippa Dudley; Ryan Greenwood; Paul D. Johnson; Jason Grant Kettle; Clare Lane; Gillian M. Lamont; Andrew G. Leach; Richard William Arthur Luke; Jeff Morris; Donald J. Ogilvie; Ken Page; Martin Pass; Stuart E. Pearson; Linette Ruston

Wide-ranging exploration of analogues of an ATP-competitive pyrrolopyrimidine inhibitor of Akt led to the discovery of clinical candidate AZD5363, which showed increased potency, reduced hERG affinity, and higher selectivity against the closely related AGC kinase ROCK. This compound demonstrated good preclinical drug metabolism and pharmacokinetics (DMPK) properties and, after oral dosing, showed pharmacodynamic knockdown of phosphorylation of Akt and downstream biomarkers in vivo, and inhibition of tumor growth in a breast cancer xenograft model.


Journal of Medicinal Chemistry | 2012

Diverse heterocyclic scaffolds as allosteric inhibitors of AKT.

Jason Grant Kettle; Simon Brown; Claire Crafter; Barry R. Davies; Phillippa Dudley; Gary Fairley; Paul Faulder; Shaun Fillery; Hannah Greenwood; Janet Hawkins; Michael Rabinder James; Keith A. Johnson; Clare Lane; Martin Pass; Jennifer H. Pink; Helen Plant; Sabina Cosulich

Wide-ranging exploration of potential replacements for a quinoline-based inhibitor of activation of AKT kinase led to number of alternative, novel scaffolds with potentially improved potency and physicochemical properties. Examples showed predictable DMPK properties, and one such compound demonstrated pharmacodynamic knockdown of phosphorylation of AKT and downstream biomarkers in vivo and inhibition of tumor growth in a breast cancer xenograft model.


Molecular Cancer Therapeutics | 2015

AZD2014, an Inhibitor of mTORC1 and mTORC2, Is Highly Effective in ER+ Breast Cancer When Administered Using Intermittent or Continuous Schedules

Sylvie Guichard; Jon Owen Curwen; Teeru Bihani; Celina D'Cruz; James W.T. Yates; Michael Grondine; Zoe Howard; Barry R. Davies; Graham Bigley; Teresa Klinowska; Kurt Gordon Pike; Martin Pass; Christine M. Chresta; Urszula M. Polanska; Robert McEwen; Oona Delpuech; Stephen Green; Sabina Cosulich

mTOR is an atypical serine threonine kinase involved in regulating major cellular functions, such as nutrients sensing, growth, and proliferation. mTOR is part of the multiprotein complexes mTORC1 and mTORC2, which have been shown to play critical yet functionally distinct roles in the regulation of cellular processes. Current clinical mTOR inhibitors only inhibit the mTORC1 complex and are derivatives of the macrolide rapamycin (rapalogs). Encouraging effects have been observed with rapalogs in estrogen receptor–positive (ER+) breast cancer patients in combination with endocrine therapy, such as aromatase inhibitors. AZD2014 is a small-molecule ATP competitive inhibitor of mTOR that inhibits both mTORC1 and mTORC2 complexes and has a greater inhibitory function against mTORC1 than the clinically approved rapalogs. Here, we demonstrate that AZD2014 has broad antiproliferative effects across multiple cell lines, including ER+ breast models with acquired resistance to hormonal therapy and cell lines with acquired resistance to rapalogs. In vivo, AZD2014 induces dose-dependent tumor growth inhibition in several xenograft and primary explant models. The antitumor activity of AZD2014 is associated with modulation of both mTORC1 and mTORC2 substrates, consistent with its mechanism of action. In combination with fulvestrant, AZD2014 induces tumor regressions when dosed continuously or using intermittent dosing schedules. The ability to dose AZD2014 intermittently, together with its ability to block signaling from both mTORC1 and mTORC2 complexes, makes this compound an ideal candidate for combining with endocrine therapies in the clinic. AZD2014 is currently in phase II clinical trials. Mol Cancer Ther; 14(11); 2508–18. ©2015 AACR.


Journal of Medicinal Chemistry | 2012

Discovery of Checkpoint Kinase Inhibitor (S)-5-(3-Fluorophenyl)-N-(piperidin-3-yl)-3-ureidothiophene-2-carboxamide (AZD7762) by Structure-Based Design and Optimization of Thiophenecarboxamide Ureas.

Oza; Susan Ashwell; Lynsie Almeida; Patrick Brassil; Jason Breed; Chun Deng; Thomas Gero; Michael Grondine; C Horn; Stephanos Ioannidis; D Liu; Paul Lyne; Nicholas John Newcombe; Martin Pass; Jon Read; S Ready; S Rowsell; Mei Su; Dorin Toader; Melissa Vasbinder; Dingwei Yu; Yan Yu; Y Xue; S Zabludoff; James W. Janetka

Checkpoint kinases CHK1 and CHK2 are activated in response to DNA damage that results in cell cycle arrest, allowing sufficient time for DNA repair. Agents that lead to abrogation of such checkpoints have potential to increase the efficacy of such compounds as chemo- and radiotherapies. Thiophenecarboxamide ureas (TCUs) were identified as inhibitors of CHK1 by high throughput screening. A structure-based approach is described using crystal structures of JNK1 and CHK1 in complex with 1 and 2 and of the CHK1-3b complex. The ribose binding pocket of CHK1 was targeted to generate inhibitors with excellent cellular potency and selectivity over CDK1and IKKβ, key features lacking from the initial compounds. Optimization of 3b resulted in the identification of a regioisomeric 3-TCU lead 12a. Optimization of 12a led to the discovery of the clinical candidate 4 (AZD7762), which strongly potentiates the efficacy of a variety of DNA-damaging agents in preclinical models.


Bioorganic & Medicinal Chemistry Letters | 2008

Discovery of a novel class of 2-ureido thiophene carboxamide checkpoint kinase inhibitors.

James W. Janetka; Lynsie Almeida; Susan Ashwell; Patrick Brassil; Kevin Daly; Chun Deng; Thomas Gero; Roberta Glynn; Candice Horn; Stephanos Ioannidis; Paul Lyne; Nicholas John Newcombe; Vibha Oza; Martin Pass; Stephanie Springer; Mei Su; Dorin Toader; Melissa Vasbinder; Dingwei Yu; Yan Yu; Sonya Zabludoff

Checkpoint kinase-1 (Chk1, CHEK1) is a Ser/Thr protein kinase that mediates the cellular response to DNA-damage. A novel class of 2-ureido thiophene carboxamide urea (TCU) Chk1 inhibitors is described. Inhibitors in this chemotype were optimized for cellular potency and selectivity over Cdk1.

Collaboration


Dive into the Martin Pass's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Udai Banerji

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge