Network


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

Hotspot


Dive into the research topics where Joseph L. Duffy is active.

Publication


Featured researches published by Joseph L. Duffy.


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.


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.


Drugs | 2012

On- and off-target pharmacology of torcetrapib: current understanding and implications for the structure activity relationships (SAR), discovery and development of cholesteryl ester-transfer protein (CETP) inhibitors.

Douglas G. Johns; Joseph L. Duffy; Timothy S. Fisher; Brian K. Hubbard; Michael J. Forrest

Lowering of serum low-density lipoprotein cholesterol (LDL-C) levels remains the primary aim of lipid management. Much progress has been made in reducing rates of cardiovascular disease morbidity and mortality, largely through increased awareness of lipid-lowering therapies and particularly through the use of high-efficacy LDL-C-lowering HMG-CoA reductase inhibitors (statins). While statins have been effective in reducing cardiovascular disease risk, many patients do not adequately achieve guideline-recommended LDL-C goals and may benefit from additional cholesterol management therapies. Low serum levels of high-density lipoprotein cholesterol (HDL-C) are considered another important determinant of cardiovascular disease risk, and increased serum HDL-C levels have been shown to be associated with reductions in the incidence of cardiovascular disease. One approach toward raising serum HDL-C levels is the inhibition of cholesteryl ester-transfer protein (CETP), a plasma protein that promotes the transfer of cholesteryl ester from HDL particles and other lipoprotein fractions to pro-atherogenic apolipoprotein B-containing lipoproteins. The inhibition of this protein raises HDL-C levels and also reduces LDL-C levels. The concept of raising HDL-C levels through pharmacological intervention of this target was validated in preclinical and clinical studies with torcetrapib, the first CETP inhibitor to be assessed inlate-stage clinical trials. The large clinical outcomes trial, ILLUMINATE, was prematurely terminated due to other unexpected pharmacological effects of torcetrapib that led to an increased risk of cardiovascular events and deaths. Thus, the ultimate effect of CETP inhibition on cardiovascular disease outcomes remains to be determined. Other CETP inhibitors currently in development do not have the adverse effects of increased blood pressure and circulating levels of aldosterone shown to be structurally related to torcetrapib. Preclinical and pharmacology studies have shown that these CETP inhibitors are distinct compared with torcetrapib and lack the features related to its off-target pharmacology. These findings indicate that the off-target activities of torcetrapib are not necessarily class effects of CETP inhibitors. Recent clinical trials have shown that dalcetrapib, anacetrapib and evacetrapib, the most advanced of these compounds in development, effectively raise HDL-C levels and lower LDL-C in the absence of off-target activities. The results of these trials are encouraging within the limits of study size and duration and provide a rationale for conducting further studies, including large clinical outcomes trials to assess whether CETP inhibition can lead to cardioprotective effects. This review summarizes the data supporting the development of CETP inhibitors as HDL-C-raising therapy, including structureactivity relationships and preclinical and clinical pharmacology studies ofknown CETP inhibitors.


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 | 2008

Discovery of imidazole carboxamides as potent and selective CCK1R agonists

Cheng Zhu; Alexa Hansen; Thomas J. Bateman; Zhesheng Chen; Tom G. Holt; James Hubert; Bindhu V. Karanam; Susan J. Lee; Jie Pan; Su Qian; Vijay Bhasker G. Reddy; Marc L. Reitman; Alison M. Strack; Vincent Tong; Drew T. Weingarth; Michael Wolff; Doug MacNeil; Ann E. Weber; Joseph L. Duffy; Scott D. Edmondson

High-throughput screening revealed diaryl pyrazole 3 as a selective albeit modest cholecystokinin 1 receptor (CCK1R) agonist. SAR studies led to the discovery and optimization of a novel class of 1,2-diaryl imidazole carboxamides. Compound 44, which was profiled extensively, showed good in vivo mouse gallbladder emptying (mGBE) and lean mouse overnight food intake (ONFI) reduction activities.


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.


Bioorganic & Medicinal Chemistry Letters | 2003

HIV protease inhibitors with picomolar potency against PI-Resistant HIV-1 by extension of the P3 substituent.

Joseph L. Duffy; Thomas A. Rano; Nancy J. Kevin; Kevin T. Chapman; William A. Schleif; David B. Olsen; Mark Stahlhut; Carrie A. Rutkowski; Lawrence C. Kuo; Lixia Jin; Jiunn H. Lin; Emilio A. Emini; James R. Tata

A biaryl pyridylfuran P(3) substituent on the hydroxyethylene isostere scaffold affords HIV protease inhibitors (PIs) with picomolar (IC(50)) potency against the protease enzymes from PI-resistant HIV-1 strains. Inclusion of a gem-dimethyl substituent afforded compound 3 with 100% oral bioavailability (dogs) and more than double the t(1/2) of indinavir. Inhibition of multiple P450 isoforms is dependent on the regiochemistry of the pyridyl nitrogen in these compounds.

Collaboration


Dive into the Joseph L. Duffy's collaboration.

Researchain Logo
Decentralizing Knowledge