Network


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

Hotspot


Dive into the research topics where Richard P. Butt is active.

Publication


Featured researches published by Richard P. Butt.


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.


Science Translational Medicine | 2016

Pharmacological reversal of a pain phenotype in iPSC-derived sensory neurons and patients with inherited erythromelalgia.

Lishuang Cao; Aoibhinn McDonnell; Anja Nitzsche; Aristos J. Alexandrou; Pierre-Philippe Saintot; Alexandre J C Loucif; Adam R Brown; Gareth T. Young; Malgorzata A. Mis; Andrew D. Randall; Stephen G. Waxman; Philip Stanley; Simon Kirby; Sanela Tarabar; Alex Gutteridge; Richard P. Butt; Ruth M. McKernan; Paul Whiting; Zahid Ali; James Bilsland; Edward B. Stevens

A selective Nav1.7 sodium channel blocker reduced hyperexcitability of iPSC-derived sensory neurons and alleviated pain in a subpopulation of patients with an inherited pain disorder. A gain in pain control Subtype-specific blockade of sodium channel Nav1.7, which is important for firing of peripheral pain-signaling neurons, is a major focus of pain research. In a new study, Cao et al. created iPSC-derived sensory neurons from patients with inherited erythromelalgia (IEM), a painful disorder in which gain-of-function Nav1.7 mutations produce hyperexcitability and hyperresponsiveness to warmth in peripheral sensory neurons. The investigators show that a new selective Nav1.7 sodium channel blocker normalized the phenotype of iPSC-derived sensory neurons carrying IEM mutations and blocked pain perception in human subjects with IEM. These results provide proof of principle that selective Nav1.7 blockade may be useful in pain alleviation. In common with other chronic pain conditions, there is an unmet clinical need in the treatment of inherited erythromelalgia (IEM). The SCN9A gene encoding the sodium channel Nav1.7 expressed in the peripheral nervous system plays a critical role in IEM. A gain-of-function mutation in this sodium channel leads to aberrant sensory neuronal activity and extreme pain, particularly in response to heat. Five patients with IEM were treated with a new potent and selective compound that blocked the Nav1.7 sodium channel resulting in a decrease in heat-induced pain in most of the patients. We derived induced pluripotent stem cell (iPSC) lines from four of five subjects and produced sensory neurons that emulated the clinical phenotype of hyperexcitability and aberrant responses to heat stimuli. When we compared the severity of the clinical phenotype with the hyperexcitability of the iPSC-derived sensory neurons, we saw a trend toward a correlation for individual mutations. The in vitro IEM phenotype was sensitive to Nav1.7 blockers, including the clinical test agent. Given the importance of peripherally expressed sodium channels in many pain conditions, our approach may have broader utility for a wide range of pain and sensory conditions.


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.


Wound Repair and Regeneration | 2006

Inhibition of procollagen C-proteinase reduces scar hypertrophy in a rabbit model of cutaneous scarring

Russell R. Reid; Jon E. Mogford; Richard P. Butt; Alex DeGiorgio‐Miller; Thomas A. Mustoe

Hypertrophic scarring, which results from excessive collagen deposition at sites of dermal wound repair, can be functionally and cosmetically debilitating to the surgical patient. Pharmacological regulation of collagen synthesis and deposition is a direct approach to the control of scar tissue formation. One of the key steps in collagen stabilization is the cleavage of the C‐terminal propeptide from the precursor molecule to form collagen fibrils, a reaction catalyzed by procollagen C‐proteinase (PCP). We tested the ability of a PCP inhibitor to reduce hypertrophic scar formation in a rabbit ear model. After the placement of four, 7‐mm dermal wounds on each ear, New Zealand white rabbits received PCP inhibitor subcutaneously in the left ear at four time points postwounding: days 7, 9, 11, 13 (early treatment; n=20 wounds) or days 11, 13, 15, 17 (late treatment; n=20 wounds). The right ear of each animal served as a control (vehicle alone). Wounds were harvested on postoperative day 28 and scar hypertrophy quantified by measurement of the scar elevation index. Early treatment of wounds with PCP inhibitor did not reduce scar formation compared with controls (p>0.05). However, late treatment resulted in a statistically significant reduction in the scar elevation index (p<0.01). Our results point not only to the potential use of PCP inhibitors to mitigate hypertrophic scarring but also to the temporal importance of drug delivery for antiscarring therapy.


British Journal of Pharmacology | 2015

A novel selective and orally bioavailable Nav1.8 channel blocker, PF‐01247324, attenuates nociception and sensory neuron excitability

Claire Elizabeth Payne; Adam R Brown; Jonathon W Theile; Alexandre J C Loucif; Aristos J. Alexandrou; Mathew D Fuller; John H. Mahoney; Brett Antonio; Aaron C. Gerlach; David Printzenhoff; Rebecca Prime; Gillian Stockbridge; Anthony J. Kirkup; Anthony W. Bannon; Steve England; Mark L. Chapman; Sharan K. Bagal; Rosemarie Roeloffs; Uma Anand; Praveen Anand; Peter J. Bungay; Mark Ian Kemp; Richard P. Butt; Edward B. Stevens

NaV1.8 ion channels have been highlighted as important molecular targets for the design of low MW blockers for the treatment of chronic pain. Here, we describe the effects of PF‐01247324, a new generation, selective, orally bioavailable Nav1.8 channel blocker of novel chemotype.


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.


PLOS ONE | 2015

Oral Administration of PF-01247324, a Subtype-Selective Nav1.8 Blocker, Reverses Cerebellar Deficits in a Mouse Model of Multiple Sclerosis

Shannon D. Shields; Richard P. Butt; Sulayman D. Dib-Hajj; Stephen G. Waxman

Cerebellar symptoms significantly diminish quality of life in patients with multiple sclerosis (MS). We previously showed that sodium channel Nav1.8, although normally restricted to peripheral somatosensory neurons, is upregulated in the cerebellum in MS, and that Nav1.8 expression is linked to ataxia and MS-like symptoms in mice. Furthermore, intracerebroventricular administration of the Nav1.8 blocker A-803467 temporarily reversed electrophysiological and behavioral manifestations of disease in a mouse MS model; unfortunately A-803467 is not orally bioavailable, diminishing the potential for translation to human patients. In the present study, we assessed the effect of per os (p.o.) dosing of a new orally bioavailable Nav1.8-selective blocker, PF-01247324, in transgenic mice expressing Nav1.8 in Purkinje neurons, and in wildtype mice in the experimental autoimmune encephalomyelitis (EAE) model. PF-01247324 was administered by oral gavage at 1000 mg/kg; control groups received an equal volume of vehicle. Behavioral assays of motor coordination, grip strength, and ataxia were performed. We observed significant improvements in motor coordination and cerebellar-like symptoms in mice that received PF-01247324 compared to control littermates that received vehicle. These preclinical proof-of-concept data suggest that PF-01247324, its derivatives, or other Nav1.8-selective blockers merit further study for providing symptomatic therapy for cerebellar dysfunction in MS and related disorders.


British Journal of Pharmacology | 2010

Estimation of binding rate constants using a simultaneous mixed‐effects method: application to monoamine transporter reuptake inhibitor reboxetine

Neil Benson; Nelleke Snelder; Bart A. Ploeger; Carolyn Napier; Harriet Sale; Nigel J.M. Birdsall; Richard P. Butt; Piet H. van der Graaf

Background and purpose:  Reboxetine is a clinically used antidepressant and is a racemic mixture of two enantiomers, SS‐ and RR‐reboxetine. The aim of the work described in this manuscript was to determine the kinetics of binding of the RR‐ and SS‐reboxetine to the human noradrenaline transporter (hNET).


British Journal of Clinical Pharmacology | 2017

The use of a battery of pain models to detect analgesic properties of compounds : a two-part four-way crossover study

Pieter Okkerse; Guido van Amerongen; Marieke L. de Kam; Jasper Stevens; Richard P. Butt; Rachel Gurrell; Albert Dahan; Joop M. A. van Gerven; Justin L. Hay; Geert Jan Groeneveld

Aim The aim was to investigate the ability of a battery of pain models to detect analgesic properties of commonly used analgesics in healthy subjects. Methods The battery consisted of tests eliciting electrical, mechanical and thermal (contact heat and cold pressor)‐pain and included a UVB model, the thermal grill illusion and a paradigm of conditioned pain modulation. Subjects were administered fentanyl 3 &mgr;g kg–1, phenytoin 300 mg, (S)‐ketamine 10 mg and placebo (part I), or imipramine 100 mg, pregabalin 300 mg, ibuprofen 600 mg and placebo (part II). Pain measurements were performed at baseline and up to 10 h post‐dose. Endpoints were analysed using a mixed model analysis of variance. Results Sixteen subjects (8 female) completed each part. The pain tolerance threshold (PTT) for electrical stimulation was increased (all P < 0.05) compared to placebo for (S)‐ketamine (+10.1%), phenytoin (+8.5%) and pregabalin (+10.8%). The PTT for mechanical pain was increased by pregabalin (+14.1%). The cold pressor PTT was increased by fentanyl (+17.1%) and pregabalin (+46.4%). Normal skin heat pain detection threshold was increased by (S)‐ketamine (+3.3%), fentanyl (+2.8%) and pregabalin (+4.1%). UVB treated skin pain detection threshold was increased by fentanyl (+2.6%) and ibuprofen (+4.0%). No differences in conditioned pain modulation were observed. Conclusion This study shows that these pain models are able to detect changes in pain thresholds after administration of different classes of analgesics in healthy subjects. The analgesic compounds all showed a unique profile in their effects on the pain tasks administered.


British Journal of Pharmacology | 2018

Pharmacology in translation: the preclinical and early clinical profile of the novel α2/3 functionally selective GABAA receptor positive allosteric modulator PF‐06372865

Sarah A. Nickolls; Rachel Gurrell; Guido van Amerongen; Juha Kammonen; Lishuang Cao; Adam R Brown; Clara Stead; Andy Mead; Christine Watson; Cathleen Hsu; Robert M. Owen; Andy Pike; Rebecca L. Fish; Laigao Chen; Ruolun Qiu; Evan D. Morris; Gang Feng; Mark Whitlock; Donal Gorman; Joop M. A. van Gerven; David S. Reynolds; Pinky Dua; Richard P. Butt

Benzodiazepines, non‐selective positive allosteric modulators (PAMs) of GABAA receptors, have significant side effects that limit their clinical utility. As many of these side effects are mediated by the α1 subunit, there has been a concerted effort to develop α2/3 subtype‐selective PAMs.

Collaboration


Dive into the Richard P. Butt's collaboration.

Top Co-Authors

Avatar

Zahid Ali

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge