Network


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

Hotspot


Dive into the research topics where Andrew M. Swensen is active.

Publication


Featured researches published by Andrew M. Swensen.


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 | 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.


Assay and Drug Development Technologies | 2010

A Pharmacologically Validated, High-Capacity, Functional Thallium Flux Assay for the Human Ether-à-go-go Related Gene Potassium Channel

William A. Schmalhofer; Andrew M. Swensen; Brande S. Thomas; John P. Felix; Rodolfo J. Haedo; Kelli Solly; Laszlo Kiss; Gregory J. Kaczorowski; Maria L. Garcia

The voltage-gated potassium channel, human Ether-à-go-go related gene (hERG), represents the molecular component of IKr, one of the potassium currents involved in cardiac action potential repolarization. Inhibition of IKr increases the duration of the ventricular action potential, reflected as a prolongation of the QT interval in the electrocardiogram, and increases the risk for potentially fatal ventricular arrhythmias. Because hERG is an appropriate surrogate for IKr, hERG assays that can identify potential safety liabilities of compounds during lead identification and optimization have been implemented. Although the gold standard for hERG evaluation is electrophysiology, this technique, even with the medium capacity, automated instruments that are currently available, does not meet the throughput demands for supporting typical medicinal chemistry efforts in the pharmaceutical environment. Assays that could provide reliable molecular pharmacology data, while operating in high capacity mode, are therefore desirable. In the present study, we describe a high-capacity, 384- and 1,536-well plate, functional thallium flux assay for the hERG channel that fulfills these criteria. This assay was optimized and validated using different structural classes of hERG inhibitors. An excellent correlation was found between the potency of these agents in the thallium flux assay and in electrophysiological recordings of channel activity using the QPatch automated patch platform. Extension of this study to include 991 medicinal chemistry compounds from different internal drug development programs indicated that the thallium flux assay was a good predictor of in vitro hERG activity. These data suggest that the hERG thallium flux assay can play an important role in supporting drug development efforts.


Journal of Pharmacology and Experimental Therapeutics | 2013

Pharmacologic Inhibition of the Renal Outer Medullary Potassium Channel Causes Diuresis and Natriuresis in the Absence of Kaliuresis

Maria L. Garcia; Birgit T. Priest; Magdalena Alonso-Galicia; Xiaoyan Zhou; John P. Felix; Brande Thomas-Fowlkes; Richard M. Brochu; Timothy Bailey; Andrew M. Swensen; Jessica Liu; Lee-Yuh Pai; Jianying Xiao; Melba Hernandez; Kimberly Hoagland; Karen Owens; Haifeng Tang; Reynalda Dejesus; Sophie Roy; Gregory J. Kaczorowski; Alexander Pasternak

The renal outer medullary potassium (ROMK) channel, which is located at the apical membrane of epithelial cells lining the thick ascending loop of Henle and cortical collecting duct, plays an important role in kidney physiology by regulating salt reabsorption. Loss-of-function mutations in the human ROMK channel are associated with antenatal type II Bartter’s syndrome, an autosomal recessive life-threatening salt-wasting disorder with mild hypokalemia. Similar observations have been reported from studies with ROMK knockout mice and rats. It is noteworthy that heterozygous carriers of Kir1.1 mutations associated with antenatal Bartter’s syndrome have reduced blood pressure and a decreased risk of developing hypertension by age 60. Although selective ROMK inhibitors would be expected to represent a new class of diuretics, this hypothesis has not been pharmacologically tested. Compound A [5-(2-(4-(2-(4-(1H-tetrazol-1-yl)phenyl)acetyl)piperazin-1-yl)ethyl)isobenzofuran-1(3H)-one)], a potent ROMK inhibitor with appropriate selectivity and characteristics for in vivo testing, has been identified. Compound A accesses the channel through the cytoplasmic side and binds to residues lining the pore within the transmembrane region below the selectivity filter. In normotensive rats and dogs, short-term oral administration of compound A caused concentration-dependent diuresis and natriuresis that were comparable to hydrochlorothiazide. Unlike hydrochlorothiazide, however, compound A did not cause any significant urinary potassium losses or changes in plasma electrolyte levels. These data indicate that pharmacologic inhibition of ROMK has the potential for affording diuretic/natriuretic efficacy similar to that of clinically used diuretics but without the dose-limiting hypokalemia associated with the use of loop and thiazide-like diuretics.


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 | 2013

Discovery of a novel sub-class of ROMK channel inhibitors typified by 5-(2-(4-(2-(4-(1H-Tetrazol-1-yl)phenyl)acetyl)piperazin-1-yl)ethyl)isobenzofuran-1(3H)-one.

Haifeng Tang; Reynald K. de Jesus; Shawn P. Walsh; Yuping Zhu; Yan Yan; Birgit T. Priest; Andrew M. Swensen; Magdalena Alonso-Galicia; John P. Felix; Richard M. Brochu; Timothy Bailey; Brande Thomas-Fowlkes; Xiaoyan Zhou; Lee-Yuh Pai; Caryn Hampton; Melba Hernandez; Karen Owens; Sophie Roy; Gregory J. Kaczorowski; Lihu Yang; Maria L. Garcia; Alexander Pasternak

A sub-class of distinct small molecule ROMK inhibitors were developed from the original lead 1. Medicinal chemistry endeavors led to novel ROMK inhibitors with good ROMK functional potency and improved hERG selectivity. Two of the described ROMK inhibitors were characterized for the first in vivo proof-of-concept biology studies, and results from an acute rat diuresis model confirmed the hypothesis that ROMK inhibitors represent new mechanism diuretic and natriuretic agents.


ACS Medicinal Chemistry Letters | 2015

Discovery of a Potent and Selective ROMK Inhibitor with Pharmacokinetic Properties Suitable for Preclinical Evaluation

Shawn P. Walsh; Aurash Shahripour; Haifeng Tang; Nardos Teumelsan; Jessica Frie; Yuping Zhu; Birgit T. Priest; Andrew M. Swensen; Jessica Liu; Michael Margulis; Richard Visconti; Adam B. Weinglass; John P. Felix; Richard M. Brochu; Timothy Bailey; Brande Thomas-Fowlkes; Magdalena Alonso-Galicia; Xiaoyan Zhou; Lee-Yuh Pai; Aaron Corona; Caryn Hampton; Melba Hernandez; Ross Bentley; Jing Chen; Kashmira Shah; Joseph M. Metzger; Michael J. Forrest; Karen Owens; Vincent Tong; Sookhee Ha

A new subseries of ROMK inhibitors exemplified by 28 has been developed from the initial screening hit 1. The excellent selectivity for ROMK inhibition over related ion channels and pharmacokinetic properties across preclinical species support further preclinical evaluation of 28 as a new mechanism diuretic. Robust pharmacodynamic effects in both SD rats and dogs have been demonstrated.


Journal of Neurophysiology | 2014

A mixed Ca2+ channel blocker, A-1264087, utilizes peripheral and spinal mechanisms to inhibit spinal nociceptive transmission in a rat model of neuropathic pain.

Jun Xu; Katharine L. Chu; Chang Z. Zhu; Wende Niforatos; Andrew M. Swensen; Xenia B. Searle; Lance Lee; Michael F. Jarvis; Steve McGaraughty

N-, T- and P/Q-type voltage-gated Ca(2+) channels are critical for regulating neurotransmitter release and cellular excitability and have been implicated in mediating pathological nociception. A-1264087 is a novel state-dependent blocker of N-, T- and P/Q-type channels. In the present studies, A-1264087 blocked (IC50 = 1.6 μM) rat dorsal root ganglia N-type Ca(2+) in a state-dependent fashion. A-1264087 (1, 3 and 10 mg/kg po) dose-dependently reduced mechanical allodynia in rats with a spinal nerve ligation (SNL) injury. A-1264087 (4 mg/kg iv) inhibited both spontaneous and mechanically evoked activity of spinal wide dynamic range (WDR) neurons in SNL rats but had no effect in uninjured rats. The inhibitory effect on WDR neurons remained in spinally transected SNL rats. Injection of A-1264087 (10 nmol/0.5 μl) into the spinal cord reduced both spontaneous and evoked WDR activity in SNL rats. Application of A-1264087 (300 nmol/20 μl) into the receptive field on the hindpaw attenuated evoked but not spontaneous firing of WDR neurons. Using electrical stimulation, A-1264087 (4 mg/kg iv) inhibited Aδ- and C-fiber evoked responses and after-discharge of WDR neurons in SNL rats. These effects by A-1264087 were not present in uninjured rats. A-1264087 moderately attenuated WDR neuron windup in both uninjured and SNL rats. In summary, these results indicate that A-1264087 selectively inhibited spinal nociceptive transmission in sensitized states through both peripheral and central mechanisms.

Researchain Logo
Decentralizing Knowledge