Network


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

Hotspot


Dive into the research topics where Joseph Ligutti is active.

Publication


Featured researches published by Joseph Ligutti.


Journal of Medicinal Chemistry | 2015

Engineering Potent and Selective Analogues of GpTx-1, a Tarantula Venom Peptide Antagonist of the NaV1.7 Sodium Channel

Justin K. Murray; Joseph Ligutti; Dong Liu; Anruo Zou; Leszek Poppe; Hongyan Li; Kristin L. Andrews; Bryan D. Moyer; Philippe Favreau; Reto Stöcklin; Les P. Miranda

NaV1.7 is a voltage-gated sodium ion channel implicated by human genetic evidence as a therapeutic target for the treatment of pain. Screening fractionated venom from the tarantula Grammostola porteri led to the identification of a 34-residue peptide, termed GpTx-1, with potent activity on NaV1.7 (IC50 = 10 nM) and promising selectivity against key NaV subtypes (20× and 1000× over NaV1.4 and NaV1.5, respectively). NMR structural analysis of the chemically synthesized three disulfide peptide was consistent with an inhibitory cystine knot motif. Alanine scanning of GpTx-1 revealed that residues Trp(29), Lys(31), and Phe(34) near the C-terminus are critical for potent NaV1.7 antagonist activity. Substitution of Ala for Phe at position 5 conferred 300-fold selectivity against NaV1.4. A structure-guided campaign afforded additive improvements in potency and NaV subtype selectivity, culminating in the design of [Ala5,Phe6,Leu26,Arg28]GpTx-1 with a NaV1.7 IC50 value of 1.6 nM and >1000× selectivity against NaV1.4 and NaV1.5.


Journal of Medicinal Chemistry | 2011

Identification of a potent, state-dependent inhibitor of Nav1.7 with oral efficacy in the formalin model of persistent pain.

Howard Bregman; Loren Berry; John L. Buchanan; April Chen; Bingfan Du; Elma Feric; Markus Hierl; Liyue Huang; David Immke; Brett Janosky; Danielle Johnson; Xingwen Li; Joseph Ligutti; Dong Liu; Annika B. Malmberg; David J. Matson; Jeff S. McDermott; Peter Miu; Hanh Nho Nguyen; Vinod F. Patel; Daniel Waldon; Ben Wilenkin; Xiao Mei Zheng; Anruo Zou; Erin F. DiMauro

Clinical human genetic studies have recently identified the tetrodotoxin (TTX) sensitive neuronal voltage gated sodium channel Nav1.7 (SCN9A) as a critical mediator of pain sensitization. Herein, we report structure-activity relationships for a novel series of 2,4-diaminotriazines that inhibit hNav1.7. Optimization efforts culminated in compound 52, which demonstrated pharmacokinetic properties appropriate for in vivo testing in rats. The binding site of compound 52 on Nav1.7 was determined to be distinct from that of local anesthetics. Compound 52 inhibited tetrodotoxin-sensitive sodium channels recorded from rat sensory neurons and exhibited modest selectivity against the hERG potassium channel and against cloned and native tetrodotoxin-resistant sodium channels. Upon oral administration to rats, compound 52 produced dose- and exposure-dependent efficacy in the formalin model of pain.


Analytical Chemistry | 2012

PADLOC: A Powerful Tool to Assign Disulfide Bond Connectivities in Peptides and Proteins by NMR Spectroscopy

Leszek Poppe; John O. Hui; Joseph Ligutti; Justin K. Murray; Paul D. Schnier

The determination of the disulfide bond connectivity in a peptide or protein represents a significant challenge. It is notoriously difficult to use NMR spectroscopy to assign disulfide connectivities because NMR spectra lack direct evidence for disulfide bonds. These bonds are typically inferred from three-dimensional structure calculations, which can result in ambiguous disulfide assignment. Here, we present a new NMR based methodology, in which the disulfide connectivity is obtained by applying Bayesian rules of inference to the local topology of cysteine residues. We illustrate how this approach successfully predicts the disulfide connectivity in proteins for which crystal structures are available in the protein data bank (PDB). We also demonstrate how this methodology is used with experimental NMR data for peptides with complex disulfide topologies, including hepcidin, Kalata-B1, and μ-Conotoxin KIIIA. In the case of μ-Conotoxin KIIIA, the PADLOC connectivity (1-15,2-9,4-16) differs from previously published results; additional evidence is presented demonstrating unequivocally that this newly proposed connectivity is correct.


Journal of Medicinal Chemistry | 2016

Single Residue Substitutions That Confer Voltage-Gated Sodium Ion Channel Subtype Selectivity in the NaV1.7 Inhibitory Peptide GpTx-1

Justin K. Murray; Jason Long; Anruo Zou; Joseph Ligutti; Kristin L. Andrews; Leszek Poppe; Kaustav Biswas; Bryan D. Moyer; Les P. Miranda

There is interest in the identification and optimization of new molecular entities selectively targeting ion channels of therapeutic relevance. Peptide toxins represent a rich source of pharmacology for ion channels, and we recently reported GpTx-1 analogs that inhibit NaV1.7, a voltage-gated sodium ion channel that is a compelling target for improved treatment of pain. Here we utilize multi-attribute positional scan (MAPS) analoging, combining high-throughput synthesis and electrophysiology, to interrogate the interaction of GpTx-1 with NaV1.7 and related NaV subtypes. After one round of MAPS analoging, we found novel substitutions at multiple residue positions not previously identified, specifically glutamic acid at positions 10 or 11 or lysine at position 18, that produce peptides with single digit nanomolar potency on NaV1.7 and 500-fold selectivity against off-target sodium channels. Docking studies with a NaV1.7 homology model and peptide NMR structure generated a model consistent with the key potency and selectivity modifications mapped in this work.


Bioorganic & Medicinal Chemistry Letters | 2012

Discovery and hit-to-lead optimization of pyrrolopyrimidines as potent, state-dependent Nav1.7 antagonists

Nagasree Chakka; Howie Bregman; Bingfan Du; Hanh Nho Nguyen; John L. Buchanan; Elma Feric; Joseph Ligutti; Dong Liu; Jeff S. McDermott; Anruo Zou; Erin F. DiMauro

Herein we describe the discovery, optimization, and structure-activity relationships of novel potent pyrrolopyrimidine Na(v)1.7 antagonists. Hit-to-lead SAR studies of the pyrrolopyrimidine core, head, and tail groups of the molecule led to the identification of pyrrolopyrimidine 48 as exceptionally potent Na(v)1.7 blocker with good selectivity over hERG and improved microsomal stability relative to our hit molecule and pyrazolopyrimidine 8 as a promising starting point for future optimization efforts.


Journal of Medicinal Chemistry | 2016

Application of a Parallel Synthetic Strategy in the Discovery of Biaryl Acyl Sulfonamides as Efficient and Selective NaV1.7 Inhibitors

Erin F. DiMauro; Stephen Altmann; Loren Berry; Howard Bregman; Nagasree Chakka; Margaret Y. Chu-Moyer; Elma Feric Bojic; Robert S. Foti; Robert T. Fremeau; Hua Gao; Hakan Gunaydin; Angel Guzman-Perez; Brian E. Hall; Hongbing Huang; Michael Jarosh; Thomas Kornecook; Josie Lee; Joseph Ligutti; Dong Liu; Bryan D. Moyer; Daniel Ortuno; Paul Rose; Laurie B. Schenkel; Kristin Taborn; Jean Wang; Yan Wang; Violeta Yu; Matthew Weiss

The majority of potent and selective hNaV1.7 inhibitors possess common pharmacophoric features that include a heteroaryl sulfonamide headgroup and a lipophilic aromatic tail group. Recently, reports of similar aromatic tail groups in combination with an acyl sulfonamide headgroup have emerged, with the acyl sulfonamide bestowing levels of selectivity over hNaV1.5 comparable to the heteroaryl sulfonamide. Beginning with commercially available carboxylic acids that met selected pharmacophoric requirements in the lipophilic tail, a parallel synthetic approach was applied to rapidly generate the derived acyl sulfonamides. A biaryl acyl sulfonamide hit from this library was elaborated, optimizing for potency and selectivity with attention to physicochemical properties. The resulting novel leads are potent, ligand and lipophilic efficient, and selective over hNaV1.5. Representative lead 36 demonstrates selectivity over other human NaV isoforms and good pharmacokinetics in rodents. The biaryl acyl sulfonamides reported herein may also offer ADME advantages over known heteroaryl sulfonamide inhibitors.


Bioorganic & Medicinal Chemistry Letters | 2012

The discovery of aminopyrazines as novel, potent Nav1.7 antagonists: Hit-to-lead identification and SAR

Howard Bregman; Hanh Nho Nguyen; Elma Feric; Joseph Ligutti; Dong Liu; Jeff S. McDermott; Ben Wilenkin; Anruo Zou; Liyue Huang; Xingwen Li; Erin F. DiMauro

Herein the discovery of a novel class of aminoheterocyclic Na(v)1.7 antagonists is reported. Hit compound 1 was potent but suffered from poor pharmacokinetics and selectivity. The compact structure of 1 offered a modular synthetic strategy towards a broad structure-activity relationship analysis. This analysis led to the identification of aminopyrazine 41, which had vastly improved hERG selectivity and pharmacokinetic properties.


Journal of Medicinal Chemistry | 2017

Sulfonamides as Selective NaV1.7 Inhibitors: Optimizing Potency and Pharmacokinetics While Mitigating Metabolic Liabilities

Matthew Weiss; Thomas Dineen; Isaac E. Marx; Steven Altmann; Alessandro Boezio; Howard Bregman; Margaret Y. Chu-Moyer; Erin F. DiMauro; Elma Feric Bojic; Robert S. Foti; Hua Gao; Russell Graceffa; Hakan Gunaydin; Angel Guzman-Perez; Hongbing Huang; Liyue Huang; Michael Jarosh; Thomas Kornecook; Charles Kreiman; Joseph Ligutti; Daniel S. La; Min-Hwa Jasmine Lin; Dong Liu; Bryan D. Moyer; Hanh Nho Nguyen; Emily A. Peterson; Paul Rose; Kristin Taborn; Beth D. Youngblood; Violeta Yu

Several reports have recently emerged regarding the identification of heteroarylsulfonamides as NaV1.7 inhibitors that demonstrate high levels of selectivity over other NaV isoforms. The optimization of a series of internal NaV1.7 leads that address a number of metabolic liabilities including bioactivation, PXR activation, as well as CYP3A4 induction and inhibition led to the identification of potent and selective inhibitors that demonstrated favorable pharmacokinetic profiles and were devoid of the aforementioned liabilities. The key to achieving this within a series prone to transporter-mediated clearance was the identification of a small range of optimal cLogD values and the discovery of subtle PXR SAR that was not lipophilicity dependent. This enabled the identification of compound 20, which was advanced into a target engagement pharmacodynamic model where it exhibited robust reversal of histamine-induced scratching bouts in mice.


Journal of Pharmacology and Experimental Therapeutics | 2017

Pharmacologic Characterization of AMG8379, a Potent and Selective Small Molecule Sulfonamide Antagonist of the Voltage-Gated Sodium Channel NaV1.7

Thomas Kornecook; Ruoyuan Yin; Stephen Altmann; Xuhai Be; Virginia Berry; Christopher P. Ilch; Michael Jarosh; Danielle Johnson; Josie H. Lee; Sonya G. Lehto; Joseph Ligutti; Dong Liu; Jason Luther; David J. Matson; Danny Ortuno; John Roberts; Kristin Taborn; Jinti Wang; Matthew Weiss; Violeta Yu; Dawn Zhu; Robert T. Fremeau; Bryan D. Moyer

Potent and selective antagonists of the voltage-gated sodium channel NaV1.7 represent a promising avenue for the development of new chronic pain therapies. We generated a small molecule atropisomer quinolone sulfonamide antagonist AMG8379 and a less active enantiomer AMG8380. Here we show that AMG8379 potently blocks human NaV1.7 channels with an IC50 of 8.5 nM and endogenous tetrodotoxin (TTX)-sensitive sodium channels in dorsal root ganglion (DRG) neurons with an IC50 of 3.1 nM in whole-cell patch clamp electrophysiology assays using a voltage protocol that interrogates channels in a partially inactivated state. AMG8379 was 100- to 1000-fold selective over other NaV family members, including NaV1.4 expressed in muscle and NaV1.5 expressed in the heart, as well as TTX-resistant NaV channels in DRG neurons. Using an ex vivo mouse skin-nerve preparation, AMG8379 blocked mechanically induced action potential firing in C-fibers in both a time-dependent and dose-dependent manner. AMG8379 similarly reduced the frequency of thermally induced C-fiber spiking, whereas AMG8380 affected neither mechanical nor thermal responses. In vivo target engagement of AMG8379 in mice was evaluated in multiple NaV1.7-dependent behavioral endpoints. AMG8379 dose-dependently inhibited intradermal histamine-induced scratching and intraplantar capsaicin-induced licking, and reversed UVB radiation skin burn–induced thermal hyperalgesia; notably, behavioral effects were not observed with AMG8380 at similar plasma exposure levels. AMG8379 is a potent and selective NaV1.7 inhibitor that blocks sodium current in heterologous cells as well as DRG neurons, inhibits action potential firing in peripheral nerve fibers, and exhibits pharmacodynamic effects in translatable models of both itch and pain.


Journal of Medicinal Chemistry | 2017

Sulfonamides as Selective NaV1.7 Inhibitors: Optimizing Potency, Pharmacokinetics, and Metabolic Properties to Obtain Atropisomeric Quinolinone (AM-0466) that Affords Robust in Vivo Activity

Russell Graceffa; Alessandro Boezio; Jessica Able; Steven Altmann; Loren Berry; Christiane Boezio; John R. Butler; Margaret Y. Chu-Moyer; Melanie Cooke; Erin F. DiMauro; Thomas Dineen; Elma Feric Bojic; Robert S. Foti; Robert T. Fremeau; Angel Guzman-Perez; Hua Gao; Hakan Gunaydin; Hongbing Huang; Liyue Huang; Christopher P. Ilch; Michael Jarosh; Thomas Kornecook; Charles Kreiman; Daniel S. La; Joseph Ligutti; Benjamin C. Milgram; Min-Hwa Jasmine Lin; Isaac E. Marx; Hanh Nho Nguyen; Emily A. Peterson

Because of its strong genetic validation, NaV1.7 has attracted significant interest as a target for the treatment of pain. We have previously reported on a number of structurally distinct bicyclic heteroarylsulfonamides as NaV1.7 inhibitors that demonstrate high levels of selectivity over other NaV isoforms. Herein, we report the discovery and optimization of a series of atropisomeric quinolinone sulfonamide inhibitors [ Bicyclic sulfonamide compounds as sodium channel inhibitors and their preparation . WO 2014201206, 2014 ] of NaV1.7, which demonstrate nanomolar inhibition of NaV1.7 and exhibit high levels of selectivity over other sodium channel isoforms. After optimization of metabolic and pharmacokinetic properties, including PXR activation, CYP2C9 inhibition, and CYP3A4 TDI, several compounds were advanced into in vivo target engagement and efficacy models. When tested in mice, compound 39 (AM-0466) demonstrated robust pharmacodynamic activity in a NaV1.7-dependent model of histamine-induced pruritus (itch) and additionally in a capsaicin-induced nociception model of pain without any confounding effect in open-field activity.

Collaboration


Dive into the Joseph Ligutti's collaboration.

Researchain Logo
Decentralizing Knowledge