Network


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

Hotspot


Dive into the research topics where Vivien A. Warren is active.

Publication


Featured researches published by Vivien A. Warren.


Journal of Pharmacology and Experimental Therapeutics | 2010

Analgesic Effects of a Substituted N-Triazole Oxindole (TROX-1), a State-Dependent, Voltage-Gated Calcium Channel 2 Blocker

Catherine Abbadie; Owen B. McManus; Shu-Yu Sun; Randal M. Bugianesi; Ge Dai; Rodolfo J. Haedo; James B Herrington; Gregory J. Kaczorowski; McHardy M. Smith; Andrew M. Swensen; Vivien A. Warren; Brande S. Williams; Stephen P. Arneric; Cyrus Eduljee; Terrance P. Snutch; Elizabeth W. Tringham; Nina Jochnowitz; Annie Liang; D. Euan MacIntyre; Erin McGowan; Shruti Mistry; Valerie V. White; Scott B. Hoyt; Clare London; Kathryn A. Lyons; Patricia B. Bunting; Sylvia Volksdorf; Joseph L. Duffy

Voltage-gated calcium channel (Cav)2.2 (N-type calcium channels) are key components in nociceptive transmission pathways. Ziconotide, a state-independent peptide inhibitor of Cav2.2 channels, is efficacious in treating refractory pain but exhibits a narrow therapeutic window and must be administered intrathecally. We have discovered an N-triazole oxindole, (3R)-5-(3-chloro-4-fluorophenyl)-3-methyl-3-(pyrimidin-5-ylmethyl)-1-(1H-1,2,4-triazol-3-yl)-1,3-dihydro-2H-indol-2-one (TROX-1), as a small-molecule, state-dependent blocker of Cav2 channels, and we investigated the therapeutic advantages of this compound for analgesia. TROX-1 preferentially inhibited potassium-triggered calcium influx through recombinant Cav2.2 channels under depolarized conditions (IC50 = 0.27 μM) compared with hyperpolarized conditions (IC50 > 20 μM). In rat dorsal root ganglion (DRG) neurons, TROX-1 inhibited ω-conotoxin GVIA-sensitive calcium currents (Cav2.2 channel currents), with greater potency under depolarized conditions (IC50 = 0.4 μM) than under hyperpolarized conditions (IC50 = 2.6 μM), indicating state-dependent Cav2.2 channel block of native as well as recombinant channels. TROX-1 fully blocked calcium influx mediated by a mixture of Cav2 channels in calcium imaging experiments in rat DRG neurons, indicating additional block of all Cav2 family channels. TROX-1 reversed inflammatory-induced hyperalgesia with maximal effects equivalent to nonsteroidal anti-inflammatory drugs, and it reversed nerve injury-induced allodynia to the same extent as pregabalin and duloxetine. In contrast, no significant reversal of hyperalgesia was observed in Cav2.2 gene-deleted mice. Mild impairment of motor function in the Rotarod test and cardiovascular functions were observed at 20- to 40-fold higher plasma concentrations than required for analgesic activities. TROX-1 demonstrates that an orally available state-dependent Cav2 channel blocker may achieve a therapeutic window suitable for the treatment of chronic pain.


Assay and Drug Development Technologies | 2004

Functional assay of voltage-gated sodium channels using membrane potential-sensitive dyes.

John P. Felix; Brande S. Williams; Birgit T. Priest; Richard M. Brochu; Ivy E. Dick; Vivien A. Warren; Lizhen Yan; Robert S. Slaughter; Gregory J. Kaczorowski; McHardy M. Smith; Maria L. Garcia

The discovery of novel therapeutic agents that act on voltage-gated sodium channels requires the establishment of high-capacity screening assays that can reliably measure the activity of these proteins. Fluorescence resonance energy transfer (FRET) technology using membrane potential-sensitive dyes has been shown to provide a readout of voltage-gated sodium channel activity in stably transfected cell lines. Due to the inherent rapid inactivation of sodium channels, these assays require the presence of a channel activator to prolong channel opening. Because sodium channel activators and test compounds may share related binding sites on the protein, the assay protocol is critical for the proper identification of channel inhibitors. In this study, high throughput, functional assays for the voltage-gated sodium channels, hNa(V)1.5 and hNa(V)1.7, are described. In these assays, channels stably expressed in HEK cells are preincubated with test compound in physiological medium and then exposed to a sodium channel activator that slows channel inactivation. Sodium ion movement through open channels causes membrane depolarization that can be measured with a FRET dye membrane potential-sensing system, providing a large and reproducible signal. Unlike previous assays, the signal obtained in the agonist initiation assay is sensitive to all sodium channel modulators that were tested and can be used in high throughput mode, as well as in support of Medicinal Chemistry efforts for lead optimization.


Assay and Drug Development Technologies | 2008

A high-throughput assay for evaluating state dependence and subtype selectivity of Cav2 calcium channel inhibitors.

Ge Dai; Rodolfo J. Haedo; Vivien A. Warren; Kevin S. Ratliff; Randal M. Bugianesi; Alison Rush; Mark E. Williams; James B Herrington; McHardy M. Smith; Owen B. McManus; Andrew M. Swensen

Cav2.2 channels play a critical role in pain signaling by controlling synaptic transmission between dorsal root ganglion neurons and dorsal horn neurons. The Cav2.2-selective peptide blocker ziconotide (Prialt, Elan Pharmaceuticals, Dublin, Ireland) has proven efficacious in pain relief, but has a poor therapeutic index and requires intrathecal administration. This has provided impetus for finding an orally active, state-dependent Cav2.2 inhibitor with an improved safety profile. Members of the Cav2 subfamily of calcium channels are the main contributors to central and peripheral synaptic transmission, but the pharmacological effects of blocking each subtype is not yet defined. Here we describe a high-throughput fluorescent assay using a fluorometric imaging plate reader (FLIPR [Molecular Devices, Sunnyvale, CA]) designed to quickly evaluate the state dependence and selectivity of inhibitors across the Cav2 subfamily. Stable cell lines expressing functional Cav2 channels (Ca(V)alpha, beta(3), and alpha(2)delta subunits) were co-transfected with an inward rectifier (Kir2.3) so that membrane potential, and therefore channel state, could be controlled by external potassium concentration. Following cell incubation in drug with varying concentrations of potassium, a high potassium trigger was added to elicit calcium influx through available, unblocked channels. State-dependent inhibitors that preferentially bind to channels in the open or inactivated state can be identified by their increased potency at higher potassium concentrations, where cells are depolarized and channels are biased towards these states. Although the Cav2 channel subtypes differ in their voltage dependence of inactivation, by adjusting pre-trigger potassium concentrations, the degree of steady-state inactivation can be more closely matched across Cav2 subtypes to assess molecular selectivity.


Molecular Pharmacology | 2012

Characterization of the Substituted N-Triazole Oxindole TROX-1, a Small-Molecule, State-Dependent Inhibitor of Cav2 Calcium Channels

Andrew M. Swensen; James B Herrington; Randal M. Bugianesi; Ge Dai; Rodolfo J. Haedo; Kevin S. Ratliff; McHardy M. Smith; Vivien A. Warren; Stephen P. Arneric; Cyrus Eduljee; David Parker; Terrance P. Snutch; Scott B. Hoyt; Clare London; Joseph L. Duffy; Gregory J. Kaczorowski; Owen B. McManus

Biological, genetic, and clinical evidence provide validation for N-type calcium channels (CaV2.2) as therapeutic targets for chronic pain. A state-dependent CaV2.2 inhibitor may provide an improved therapeutic window over ziconotide, the peptidyl CaV2.2 inhibitor used clinically. Supporting this notion, we recently reported that in preclinical models, the state-dependent CaV2 inhibitor (3R)-5-(3-chloro-4-fluorophenyl)-3-methyl-3-(pyrimidin-5-ylmethyl)-1-(1H-1,2,4-triazol-3-yl)-1,3-dihydro-2H-indol-2-one (TROX-1) has an improved therapeutic window compared with ziconotide. Here we characterize TROX-1 inhibition of Cav2.2 channels in more detail. When channels are biased toward open/inactivated states by depolarizing the membrane potential under voltage-clamp electrophysiology, TROX-1 inhibits CaV2.2 channels with an IC50 of 0.11 μM. The voltage dependence of CaV2.2 inhibition was examined using automated electrophysiology. TROX-1 IC50 values were 4.2, 0.90, and 0.36 μM at −110, −90, and −70 mV, respectively. TROX-1 displayed use-dependent inhibition of CaV2.2 with a 10-fold IC50 separation between first (27 μM) and last (2.7 μM) pulses in a train. In a fluorescence-based calcium influx assay, TROX-1 inhibited CaV2.2 channels with an IC50 of 9.5 μM under hyperpolarized conditions and 0.69 μM under depolarized conditions. Finally, TROX-1 potency was examined across the CaV2 subfamily. Depolarized IC50 values were 0.29, 0.19, and 0.28 μM by manual electrophysiology using matched conditions and 1.8, 0.69, and 1.1 μM by calcium influx for CaV2.1, CaV2.2, and CaV2.3, respectively. Together, these in vitro data support the idea that a state-dependent, non–subtype-selective CaV2 channel inhibitor can achieve an improved therapeutic window over the relatively state-independent CaV2.2-selective inhibitor ziconotide in preclinical models of chronic pain.


Bioorganic & Medicinal Chemistry Letters | 2008

Imidazopyridines: a novel class of hNav1.7 channel blockers.

Clare London; Scott B. Hoyt; William H. Parsons; Brande S. Williams; Vivien A. Warren; Richard Tschirret-Guth; McHardy M. Smith; Birgit T. Priest; Erin McGowan; William J. Martin; Kathryn A. Lyons; Xiaohua Li; Bindhu V. Karanam; Nina Jochnowitz; Maria L. Garcia; John P. Felix; Brian Dean; Catherine Abbadie; Gregory J. Kaczorowski; Joseph L. Duffy

A series of imidazopyridines were evaluated as potential sodium channel blockers for the treatment of neuropathic pain. Several members were identified with good hNa(v)1.7 potency and excellent rat pharmacokinetic profiles. Compound 4 had good efficacy (52% and 41% reversal of allodynia at 2 and 4h post-dose, respectively) in the Chung rat spinal nerve ligation (SNL) model of neuropathic pain when dosed orally at 10mg/kg.


Bioorganic & Medicinal Chemistry Letters | 2011

A potent and selective indole N-type calcium channel (Cav2.2) blocker for the treatment of pain

Sriram Tyagarajan; Prasun K. Chakravarty; Min Park; Bishan Zhou; James B Herrington; Kevin S. Ratliff; Randall M. Bugianesi; Brande S. Williams; Rodolfo J. Haedo; Andrew M. Swensen; Vivien A. Warren; McHardy M. Smith; Maria L. Garcia; Gregory J. Kaczorowski; Owen B. McManus; Kathryn A. Lyons; Xiaohua Li; Maria Madeira; Bindhu V. Karanam; Mitchell D. Green; Michael J. Forrest; Catherine Abbadie; Erin McGowan; Shruti Mistry; Nina Jochnowitz; Joseph L. Duffy

N-type calcium channels (Ca(v)2.2) have been shown to play a critical role in pain. A series of low molecular weight 2-aryl indoles were identified as potent Ca(v)2.2 blockers with good in vitro and in vivo potency.


Journal of Medicinal Chemistry | 2012

Aminopiperidine sulfonamide Cav2.2 channel inhibitors for the treatment of chronic pain.

Pengcheng P. Shao; Feng Ye; Prasun K. Chakravarty; Deepu J. Varughese; James B Herrington; Ge Dai; Randal M. Bugianesi; Rodolfo J. Haedo; Andrew M. Swensen; Vivien A. Warren; McHardy M. Smith; Maria L. Garcia; Owen B. McManus; Kathryn A. Lyons; Xiaohua Li; Mitchell D. Green; Nina Jochnowitz; Erin McGowan; Shruti Mistry; Shu-Yu Sun; Catherine Abbadie; Gregory J. Kaczorowski; Joseph L. Duffy

The voltage-gated calcium channel Ca(v)2.2 (N-type calcium channel) is a critical regulator of synaptic transmission and has emerged as an attractive target for the treatment of chronic pain. We report here the discovery of sulfonamide-derived, state-dependent inhibitors of Ca(v)2.2. In particular, 19 is an inhibitor of Ca(v)2.2 that is selective over cardiac ion channels, with a good preclinical PK and biodistribution profile. This compound exhibits dose-dependent efficacy in preclinical models of inflammatory hyperalgesia and neuropathic allodynia and is devoid of ancillary cardiovascular or CNS pharmacology at the doses tested. Importantly, 19 exhibited no efficacy in Ca(v)2.2 gene-deleted mice. The discovery of metabolite 26 confounds further development of members of this aminopiperidine sulfonamide series. This discovery also suggests specific structural liabilities of this class of compounds that must be addressed.


Bioorganic & Medicinal Chemistry Letters | 2010

Discovery of a novel class of biphenyl pyrazole sodium channel blockers for treatment of neuropathic pain.

Sriram Tyagarajan; Prasun K. Chakravarty; Bishan Zhou; Brett Taylor; Ronsar Eid; Michael H. Fisher; William H. Parsons; Mathew J. Wyvratt; Kathryn A. Lyons; Tracy Klatt; Xiaohua Li; Sanjeev Kumar; Brande S. Williams; John P. Felix; Birgit T. Priest; Richard M. Brochu; Vivien A. Warren; McHardy M. Smith; Maria L. Garcia; Gregory J. Kaczorowski; William J. Martin; Catherine Abbadie; Erin McGowan; Nina Jochnowitz; Ann E. Weber; Joseph L. Duffy

A series of novel biphenyl pyrazole dicarboxamides were identified as potential sodium channel blockers for treatment of neuropathic pain. Compound 20 had outstanding efficacy in the Chung rat spinal nerve ligation (SNL) model of neuropathic pain.


Bioorganic & Medicinal Chemistry Letters | 2010

Substituted biaryl oxazoles, imidazoles, and thiazoles as sodium channel blockers.

Sriram Tyagarajan; Prasun K. Chakravarty; Bishan Zhou; Michael H. Fisher; Mathew J. Wyvratt; Kathy Lyons; Tracy Klatt; Xiaohua Li; Sanjeev Kumar; Brande S. Williams; John P. Felix; Birgit T. Priest; Richard M. Brochu; Vivien A. Warren; McHardy M. Smith; Maria L. Garcia; Gregory J. Kaczorowski; William J. Martin; Catherine Abbadie; Erin McGowan; Nina Jochnowitz; William H. Parsons

Voltage-gated sodium channels have been shown to play a critical role in neuropathic pain. With a goal to develop potent peripherally active sodium channel blockers, a series of low molecular weight biaryl substituted imidazoles, oxazoles, and thiazole carboxamides were identified with good in vitro and in vivo potency.


Bioorganic & Medicinal Chemistry Letters | 2013

A novel benzazepinone sodium channel blocker with oral efficacy in a rat model of neuropathic pain

Scott B. Hoyt; Clare London; Catherine Abbadie; John P. Felix; Maria L. Garcia; Nina Jochnowitz; Bindhu V. Karanam; Xiaohua Li; Kathryn A. Lyons; Erin McGowan; Birgit T. Priest; McHardy M. Smith; Vivien A. Warren; Brande Thomas-Fowlkes; Gregory J. Kaczorowski; Joseph L. Duffy

A series of benzazepinones were synthesized and evaluated for block of Nav1.7 sodium channels. Compound 30 from this series displayed potent channel block, good selectivity versus other targets, and dose-dependent oral efficacy in a rat model of neuropathic pain.

Collaboration


Dive into the Vivien A. Warren's collaboration.

Researchain Logo
Decentralizing Knowledge