Network


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

Hotspot


Dive into the research topics where Brian Edward Marron is active.

Publication


Featured researches published by Brian Edward Marron.


Proceedings of the National Academy of Sciences of the United States of America | 2007

A-803467, a potent and selective Nav1.8 sodium channel blocker, attenuates neuropathic and inflammatory pain in the rat.

Michael F. Jarvis; Prisca Honore; Char Chang Shieh; Mark L. Chapman; Shailen K. Joshi; Xu Feng Zhang; Michael E. Kort; William L. Carroll; Brian Edward Marron; Robert Nelson Atkinson; James P. Thomas; Dong Liu; Michael J. Krambis; Yi Liu; Steve McGaraughty; Katharine L. Chu; Rosemarie Roeloffs; Chengmin Zhong; Joseph P. Mikusa; Gricelda Hernandez; Donna M. Gauvin; Carrie L. Wade; Chang Zhu; Madhavi Pai; Marc Scanio; Lei Shi; Irene Drizin; Robert J. Gregg; Mark A. Matulenko; Ahmed A. Hakeem

Activation of tetrodotoxin-resistant sodium channels contributes to action potential electrogenesis in neurons. Antisense oligonucleotide studies directed against Nav1.8 have shown that this channel contributes to experimental inflammatory and neuropathic pain. We report here the discovery of A-803467, a sodium channel blocker that potently blocks tetrodotoxin-resistant currents (IC50 = 140 nM) and the generation of spontaneous and electrically evoked action potentials in vitro in rat dorsal root ganglion neurons. In recombinant cell lines, A-803467 potently blocked human Nav1.8 (IC50 = 8 nM) and was >100-fold selective vs. human Nav1.2, Nav1.3, Nav1.5, and Nav1.7 (IC50 values ≥1 μM). A-803467 (20 mg/kg, i.v.) blocked mechanically evoked firing of wide dynamic range neurons in the rat spinal dorsal horn. A-803467 also dose-dependently reduced mechanical allodynia in a variety of rat pain models including: spinal nerve ligation (ED50 = 47 mg/kg, i.p.), sciatic nerve injury (ED50 = 85 mg/kg, i.p.), capsaicin-induced secondary mechanical allodynia (ED50 ≈ 100 mg/kg, i.p.), and thermal hyperalgesia after intraplantar complete Freunds adjuvant injection (ED50 = 41 mg/kg, i.p.). A-803467 was inactive against formalin-induced nociception and acute thermal and postoperative pain. These data demonstrate that acute and selective pharmacological blockade of Nav1.8 sodium channels in vivo produces significant antinociception in animal models of neuropathic and inflammatory pain.


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 | 2008

Discovery and biological evaluation of 5-aryl-2-furfuramides, potent and selective blockers of the Nav1.8 sodium channel with efficacy in models of neuropathic and inflammatory pain.

Michael E. Kort; Irene Drizin; Robert J. Gregg; Marc Scanio; Lei Shi; Michael F. Gross; Robert Nelson Atkinson; Matthew S. Johnson; Gregory J. Pacofsky; James B. Thomas; William A. Carroll; Michael J. Krambis; Dong Liu; Char-Chang Shieh; Xu-Feng Zhang; Gricelda Hernandez; Joseph P. Mikusa; Chengmin Zhong; Shailen K. Joshi; Prisca Honore; Rosemarie Roeloffs; Kennan C. Marsh; Bernard P. Murray; Jinrong Liu; Stephen Werness; Connie R. Faltynek; Douglas S. Krafte; Michael F. Jarvis; Mark L. Chapman; Brian Edward Marron

Nav1.8 (also known as PN3) is a tetrodotoxin-resistant (TTx-r) voltage-gated sodium channel (VGSC) that is highly expressed on small diameter sensory neurons and has been implicated in the pathophysiology of inflammatory and neuropathic pain. Recent studies using an Nav1.8 antisense oligonucleotide in an animal model of chronic pain indicated that selective blockade of Nav1.8 was analgesic and could provide effective analgesia with a reduction in the adverse events associated with nonselective VGSC blocking therapeutic agents. Herein, we describe the preparation and characterization of a series of 5-substituted 2-furfuramides, which are potent, voltage-dependent blockers (IC50 < 10 nM) of the human Nav1.8 channel. Selected derivatives, such as 7 and 27, also blocked TTx-r sodium currents in rat dorsal root ganglia (DRG) neurons with comparable potency and displayed >100-fold selectivity versus human sodium (Nav1.2, Nav1.5, Nav1.7) and human ether-a-go-go (hERG) channels. Following systemic administration, compounds 7 and 27 dose-dependently reduced neuropathic and inflammatory pain in experimental rodent models.


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.


Neuropharmacology | 2010

A-887826 is a structurally novel, potent and voltage-dependent Nav1.8 sodium channel blocker that attenuates neuropathic tactile allodynia in rats

Xu-Feng Zhang; Char-Chang Shieh; Mark L. Chapman; Mark A. Matulenko; Ahmed H. Hakeem; Robert N. Atkinson; Michael E. Kort; Brian Edward Marron; Shailen K. Joshi; Prisca Honore; Connie R. Faltynek; Douglas S. Krafte; Michael F. Jarvis

Activation of sodium channels is essential to action potential generation and propagation. Recent genetic and pharmacological evidence indicates that activation of Na(v)1.8 channels contributes to chronic pain. Herein, we describe the identification of a novel series of structurally related pyridine derivatives as potent Na(v)1.8 channel blockers. A-887826 exemplifies this series and potently (IC(50)=11nM) blocked recombinant human Na(v)1.8 channels. A-887826 was approximately 3 fold less potent to block Na(v)1.2, approximately 10 fold less potent to block tetrodotoxin-sensitive sodium (TTX-S Na(+)) currents and was >30 fold less potent to block Na(V)1.5 channels. A-887826 potently blocked tetrodotoxin-resistant sodium (TTX-R Na(+)) currents (IC(50)=8nM) from small diameter rat dorsal root ganglion (DRG) neurons in a voltage-dependent fashion. A-887826 effectively suppressed evoked action potential firing when DRG neurons were held at depolarized potentials and reversibly suppressed spontaneous firing in small diameter DRG neurons from complete Freunds adjuvant inflamed rats. Following oral administration, A-887826 significantly attenuated tactile allodynia in a rat neuropathic pain model. Further characterization of TTX-R current block in rat DRG neurons demonstrated that A-887826 (100nM) shifted the mid-point of voltage-dependent inactivation of TTX-R currents by approximately 4mV without affecting voltage-dependent activation and did not exhibit frequency-dependent inhibition. The present data demonstrate that A-887826 is a structurally novel and potent Na(v)1.8 blocker that inhibits rat DRG TTX-R currents in a voltage-, but not frequency-dependent fashion. The ability of this structurally novel Na(v)1.8 blocker to effectively reduce tactile allodynia in neuropathic rats further supports the role of Na(v)1.8 sodium channels in pathological pain states.


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 | 2010

Discovery and biological evaluation of potent, selective, orally bioavailable, pyrazine-based blockers of the Nav1.8 sodium channel with efficacy in a model of neuropathic pain

Marc Scanio; Lei Shi; Irene Drizin; Robert J. Gregg; Robert N. Atkinson; James B. Thomas; Matthew Johnson; Mark L. Chapman; Dong Liu; Michael J. Krambis; Yi Liu; Char-Chang Shieh; Xu-Feng Zhang; Gricelda Simler; Shailen K. Joshi; Prisca Honore; Kennan C. Marsh; Alison Knox; Stephen Werness; Brett Antonio; Douglas S. Krafte; Michael F. Jarvis; Connie R. Faltynek; Brian Edward Marron; Michael E. Kort

Na(v)1.8 (also known as PN3) is a tetrodotoxin-resistant (TTx-r) voltage-gated sodium channel (VGSC) that is highly expressed on small diameter sensory neurons. It has been implicated in the pathophysiology of inflammatory and neuropathic pain, and we envisioned that selective blockade of Na(v)1.8 would be analgesic, while reducing adverse events typically associated with non-selective VGSC blocking therapeutic agents. Herein, we describe the preparation and characterization of a series of 6-aryl-2-pyrazinecarboxamides, which are potent blockers of the human Na(v)1.8 channel and also block TTx-r sodium currents in rat dorsal root ganglia (DRG) neurons. Selected derivatives display selectivity versus human Na(v)1.2. We further demonstrate that an example from this series is orally bioavailable and produces antinociceptive activity in vivo in a rodent model of neuropathic pain following oral administration.


Bioorganic & Medicinal Chemistry Letters | 2010

Subtype-selective Nav1.8 sodium channel blockers: Identification of potent, orally active nicotinamide derivatives

Michael E. Kort; Robert N. Atkinson; James B. Thomas; Irene Drizin; Matthew Johnson; Matthew A. Secrest; Robert J. Gregg; Marc Scanio; Lei Shi; Ahmed H. Hakeem; Mark A. Matulenko; Mark L. Chapman; Michael J. Krambis; Dong Liu; Char-Chang Shieh; Xu-Feng Zhang; Gricelda Simler; Joseph P. Mikusa; Chengmin Zhong; Shailen K. Joshi; Prisca Honore; Rosemarie Roeloffs; Stephen Werness; Brett Antonio; Kennan C. Marsh; Connie R. Faltynek; Douglas S. Krafte; Michael F. Jarvis; Brian Edward Marron

A series of aryl-substituted nicotinamide derivatives with selective inhibitory activity against the Na(v)1.8 sodium channel is reported. Replacement of the furan nucleus and homologation of the anilide linker in subtype-selective blocker A-803467 (1) provided potent, selective derivatives with improved aqueous solubility and oral bioavailability. Representative compounds from this series displayed efficacy in rat models of inflammatory and neuropathic pain.


Bioorganic & Medicinal Chemistry | 2008

Discovery of potent furan piperazine sodium channel blockers for treatment of neuropathic pain

Irene Drizin; Robert J. Gregg; Marc Scanio; Lei Shi; Michael Francis Gross; Robert N. Atkinson; James B. Thomas; Matthew Johnson; William A. Carroll; Brian Edward Marron; Mark L. Chapman; Dong Liu; Michael J. Krambis; Char-Chang Shieh; Xu-Feng Zhang; Gricelda Hernandez; Donna M. Gauvin; Joseph P. Mikusa; Chang Z. Zhu; Shailen K. Joshi; Prisca Honore; Kennan C. Marsh; Rosemarie Roeloffs; Stephen Werness; Douglas S. Krafte; Michael F. Jarvis; Connie R. Faltynek; Michael E. Kort

The synthesis and pharmacological characterization of a novel furan-based class of voltage-gated sodium channel blockers is reported. Compounds were evaluated for their ability to block the tetrodotoxin-resistant sodium channel Na(v)1.8 (PN3) as well as the Na(v)1.2 and Na(v)1.5 subtypes. Benchmark compounds from this series possessed enhanced potency, oral bioavailability, and robust efficacy in a rodent model of neuropathic pain, together with improved CNS and cardiovascular safety profiles compared to the clinically used sodium channel blockers mexiletine and lamotrigine.

Collaboration


Dive into the Brian Edward Marron's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael E. Kort

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge