Network


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

Hotspot


Dive into the research topics where Nigel Alan Swain is active.

Publication


Featured researches published by Nigel Alan Swain.


Journal of Medicinal Chemistry | 2013

Ion Channels as Therapeutic Targets: A Drug Discovery Perspective

Sharan K. Bagal; Alan Daniel Brown; Peter J. Cox; Kiyoyuki Omoto; Robert M. Owen; David C. Pryde; Benjamin Sidders; Sarah Elizabeth Skerratt; Edward B. Stevens; R. Ian Storer; Nigel Alan Swain

Ion channels are membrane proteins expressed in almost all living cells. The sequencing of the human genome has identified more than 400 putative ion channels, but only a fraction of these have been cloned and functionally tested. The widespread tissue distribution of ion channels, coupled with the plethora of physiological consequences of their opening and closing, makes ion-channel-targeted drug discovery highly compelling. However, despite some important drugs in clinical use today, as a class, ion channels remain underexploited in drug discovery and many existing drugs are poorly selective with significant toxicities or suboptimal efficacy. This Perspective seeks to review the ion channel family, its structural and functional features, and the diseases that are known to be modulated by members of the family. In particular, we will explore the structure and properties of known ligands and consider the future prospects for drug discovery in this challenging but high potential area.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Voltage sensor interaction site for selective small molecule inhibitors of voltage-gated sodium channels

Ken McCormack; Sonia Santos; Mark L. Chapman; Douglas S. Krafte; Brian Edward Marron; Christopher William West; Michael J. Krambis; Brett M. Antonio; Shannon Zellmer; David Printzenhoff; Karen Padilla; Zhixin Lin; P. Kay Wagoner; Nigel Alan Swain; Paul Anthony Stupple; Marcel J. de Groot; Richard P. Butt; Neil A. Castle

Significance Voltage-gated sodium (Nav) channels contribute to physiological and pathophysiological electrical signaling in nerve and muscle cells. Because Nav channel isoforms exhibit tissue-specific expression, subtype selective modulation of this channel family provides important drug development opportunities. However, most available Nav channel modulators are unable to distinguish between Nav channel subtypes, which limits their therapeutic utility because of cardiac or nervous system toxicity. This study describes a new class of subtype selective Nav channel inhibitors that interact with a region of the channel that controls voltage sensitivity. This interaction site may enable development of selective therapeutic interventions with reduced potential for toxicity. Voltage-gated sodium (Nav) channels play a fundamental role in the generation and propagation of electrical impulses in excitable cells. Here we describe two unique structurally related nanomolar potent small molecule Nav channel inhibitors that exhibit up to 1,000-fold selectivity for human Nav1.3/Nav1.1 (ICA-121431, IC50, 19 nM) or Nav1.7 (PF-04856264, IC50, 28 nM) vs. other TTX-sensitive or resistant (i.e., Nav1.5) sodium channels. Using both chimeras and single point mutations, we demonstrate that this unique class of sodium channel inhibitor interacts with the S1–S4 voltage sensor segment of homologous Domain 4. Amino acid residues in the “extracellular” facing regions of the S2 and S3 transmembrane segments of Nav1.3 and Nav1.7 seem to be major determinants of Nav subtype selectivity and to confer differences in species sensitivity to these inhibitors. The unique interaction region on the Domain 4 voltage sensor segment is distinct from the structural domains forming the channel pore, as well as previously characterized interaction sites for other small molecule inhibitors, including local anesthetics and TTX. However, this interaction region does include at least one amino acid residue [E1559 (Nav1.3)/D1586 (Nav1.7)] that is important for Site 3 α-scorpion and anemone polypeptide toxin modulators of Nav channel inactivation. The present study provides a potential framework for identifying subtype selective small molecule sodium channel inhibitors targeting interaction sites away from the pore region.


Bioorganic & Medicinal Chemistry Letters | 2014

Recent progress in sodium channel modulators for pain

Sharan K. Bagal; Mark L. Chapman; Brian Edward Marron; Rebecca Prime; R. Ian Storer; Nigel Alan Swain

Voltage-gated sodium channels (Navs) are an important family of transmembrane ion channel proteins and Nav drug discovery is an exciting field. Pharmaceutical investment in Navs for pain therapeutics has expanded exponentially due to genetic data such as SCN10A mutations and an improved ability to establish an effective screen sequence for example IonWorks Barracuda®, Synchropatch® and Qube®. Moreover, emerging clinical data (AZD-3161, XEN402, CNV1014802, PF-05089771, PF-04531083) combined with recent breakthroughs in Nav structural biology pave the way for a future of fruitful prospective Nav drug discovery.


Journal of Medicinal Chemistry | 2009

Novel Indazole Non-Nucleoside Reverse Transcriptase Inhibitors Using Molecular Hybridization Based on Crystallographic Overlays

Lyn H. Jones; Gill Allan; Oscar Barba; Catherine Burt; Romuald Corbau; Thomas Dupont; Thorsten Knöchel; Steve Irving; Donald Stuart Middleton; Charles Eric Mowbray; Manos Perros; Heather Ringrose; Nigel Alan Swain; Robert G. Webster; Mike Westby; Christopher Phillips

A major problem associated with non-nucleoside reverse transcriptase inhibitors (NNRTIs) for the treatment of HIV is their lack of resilience to mutations in the reverse transcriptase (RT) enzyme. Using structural overlays of the known inhibitors efavirenz and capravirine complexed in RT as a starting point, and structure-based drug design techniques, we have created a novel series of indazole NNRTIs that possess excellent metabolic stability and mutant resilience.


PLOS ONE | 2016

Subtype-Selective Small Molecule Inhibitors Reveal a Fundamental Role for Nav1.7 in Nociceptor Electrogenesis, Axonal Conduction and Presynaptic Release

Aristos J. Alexandrou; Adam R Brown; Mark L. Chapman; Mark Estacion; Jamie Turner; Malgorzata A. Mis; Anna Wilbrey; Elizabeth C. Payne; Alex Gutteridge; Peter Cox; Rachel Doyle; David Printzenhoff; Zhixin Lin; Brian Edward Marron; Christopher L West; Nigel Alan Swain; R. Ian Storer; Paul Anthony Stupple; Neil A. Castle; James A. Hounshell; Mirko Rivara; Andrew D. Randall; Sulayman D. Dib-Hajj; Douglas S. Krafte; Stephen G. Waxman; Manoj K. Patel; Richard P. Butt; Edward B. Stevens

Human genetic studies show that the voltage gated sodium channel 1.7 (Nav1.7) is a key molecular determinant of pain sensation. However, defining the Nav1.7 contribution to nociceptive signalling has been hampered by a lack of selective inhibitors. Here we report two potent and selective arylsulfonamide Nav1.7 inhibitors; PF-05198007 and PF-05089771, which we have used to directly interrogate Nav1.7’s role in nociceptor physiology. We report that Nav1.7 is the predominant functional TTX-sensitive Nav in mouse and human nociceptors and contributes to the initiation and the upstroke phase of the nociceptor action potential. Moreover, we confirm a role for Nav1.7 in influencing synaptic transmission in the dorsal horn of the spinal cord as well as peripheral neuropeptide release in the skin. These findings demonstrate multiple contributions of Nav1.7 to nociceptor signalling and shed new light on the relative functional contribution of this channel to peripheral and central noxious signal transmission.


Channels | 2015

Voltage gated sodium channels as drug discovery targets.

Sharanjeet Kaur Bagal; Brian Edward Marron; Robert M. Owen; Robert Ian Storer; Nigel Alan Swain

Voltage-gated sodium (NaV) channels are a family of transmembrane ion channel proteins. They function by forming a gated, water-filled pore to help establish and control cell membrane potential via control of the flow of ions between the intracellular and the extracellular environments. Blockade of NaVs has been successfully accomplished in the clinic to enable control of pathological firing patterns that occur in a diverse range of conditions such as chronic pain, epilepsy, and cardiac arrhythmias. First generation sodium channel modulator drugs, despite low inherent subtype selectivity, preferentially act on over-excited cells which reduces undesirable side effects in the clinic. However, the limited therapeutic indices observed with the first generation demanded a new generation of sodium channel inhibitors. The structure, function and the state of the art in sodium channel modulator drug discovery are discussed in this chapter.


Bioorganic & Medicinal Chemistry Letters | 2011

Challenges of drug discovery in novel target space. The Discovery and Evaluation of PF-3893787: A Novel Histamine H4 Receptor Antagonist

Charles Eric Mowbray; Andrew Simon Bell; Nick Clarke; Michelle Collins; Rhys M. Jones; Charlotte Alice Louise Lane; Wai L. Liu; Sandra D. Newman; Michael Paradowski; Emanuel Schenck; Mathew D. Selby; Nigel Alan Swain; David H. Williams

We describe the development of novel benzimidazoles as small molecule histamine H4 receptor (H4R) antagonists and their profiling in rat early toxicity studies. The discovery and optimisation of a second series of pyrimidine based antagonists is then described culminating in the identification of the clinical development candidate 13 (PF-3893787). The pre-clinical profile of 13 (PF-3893787) is presented including the development of a translatable biomarker. Our pragmatic approach to target selection, safety assessment, and testing for efficacy faced numerous challenges and we share a number of lessons which the team learned and which will assist us and others in future drug discovery projects.


MedChemComm | 2010

Aromatic chloride to nitrile transformation: medicinal and synthetic chemistry

Lyn H. Jones; Nicholas William Summerhill; Nigel Alan Swain; James E. J. Mills

This review highlights the medicinal and synthetic chemistry relevance of replacing an aromatic chloride motif with an aromatic nitrile. We explore the desirable features that this transformation can bring in a drug design sense and the recent synthetic chemistry advances that effect this replacement in a single step.


Bioorganic & Medicinal Chemistry Letters | 2012

Synthesis of novel histamine H4 receptor antagonists.

Charlotte Alice Louise Lane; Duncan Hay; Charles Eric Mowbray; Michael Paradowski; Matthew D. Selby; Nigel Alan Swain; David H. Williams

This letter describes the discovery and synthesis of a series of octahydropyrrolo[3,4-c]pyrrole based selective histamine hH4 receptor antagonists. The amidine compound 20 was found to be a potent and selective histamine H4 receptor antagonist with moderate clearance and a high volume of distribution.


Journal of Medicinal Chemistry | 2017

Discovery of Clinical Candidate 4-[2-(5-Amino-1H-pyrazol-4-yl)-4-chlorophenoxy]-5-chloro-2-fluoro-N-1,3-thiazol-4-ylbenzenesulfonamide (PF-05089771): Design and Optimization of Diaryl Ether Aryl Sulfonamides as Selective Inhibitors of NaV1.7

Nigel Alan Swain; Dave Batchelor; Serge Beaudoin; Bruce M. Bechle; Paul Anthony Bradley; Alan Daniel Brown; Bruce Brown; Kenneth John Butcher; Richard P. Butt; Mark L. Chapman; Stephen Martin Denton; David Ellis; Sebastien Rene Gabriel Galan; Stephen M Gaulier; Ben S. Greener; Marcel J. de Groot; Mel S Glossop; Ian Gurrell; Jo Hannam; Matthew S. Johnson; Zhixin Lin; Christopher John Markworth; Brian Edward Marron; David Simon Millan; Shoko Nakagawa; Andy Pike; David Printzenhoff; David James Rawson; Sarah J Ransley; Steven Reister

A series of acidic diaryl ether heterocyclic sulfonamides that are potent and subtype selective NaV1.7 inhibitors is described. Optimization of early lead matter focused on removal of structural alerts, improving metabolic stability and reducing cytochrome P450 inhibition driven drug-drug interaction concerns to deliver the desired balance of preclinical in vitro properties. Concerns over nonmetabolic routes of clearance, variable clearance in preclinical species, and subsequent low confidence human pharmacokinetic predictions led to the decision to conduct a human microdose study to determine clinical pharmacokinetics. The design strategies and results from preclinical PK and clinical human microdose PK data are described leading to the discovery of the first subtype selective NaV1.7 inhibitor clinical candidate PF-05089771 (34) which binds to a site in the voltage sensing domain.

Collaboration


Dive into the Nigel Alan Swain's collaboration.

Researchain Logo
Decentralizing Knowledge