Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Mol Pharmacol ; 105(3): 233-249, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38195157

ABSTRACT

Discovery and development of new molecules directed against validated pain targets is required to advance the treatment of pain disorders. Voltage-gated sodium channels (NaVs) are responsible for action potential initiation and transmission of pain signals. NaV1.8 is specifically expressed in peripheral nociceptors and has been genetically and pharmacologically validated as a human pain target. Selective inhibition of NaV1.8 can ameliorate pain while minimizing effects on other NaV isoforms essential for cardiac, respiratory, and central nervous system physiology. Here we present the pharmacology, interaction site, and mechanism of action of LTGO-33, a novel NaV1.8 small molecule inhibitor. LTGO-33 inhibited NaV1.8 in the nM potency range and exhibited over 600-fold selectivity against human NaV1.1-NaV1.7 and NaV1.9. Unlike prior reported NaV1.8 inhibitors that preferentially interacted with an inactivated state via the pore region, LTGO-33 was state-independent with similar potencies against closed and inactivated channels. LTGO-33 displayed species specificity for primate NaV1.8 over dog and rodent NaV1.8 and inhibited action potential firing in human dorsal root ganglia neurons. Using chimeras combined with mutagenesis, the extracellular cleft of the second voltage-sensing domain was identified as the key site required for channel inhibition. Biophysical mechanism of action studies demonstrated that LTGO-33 inhibition was relieved by membrane depolarization, suggesting the molecule stabilized the deactivated state to prevent channel opening. LTGO-33 equally inhibited wild-type and multiple NaV1.8 variants associated with human pain disorders. These collective results illustrate LTGO-33 inhibition via both a novel interaction site and mechanism of action previously undescribed in NaV1.8 small molecule pharmacologic space. SIGNIFICANCE STATEMENT: NaV1.8 sodium channels primarily expressed in peripheral pain-sensing neurons represent a validated target for the development of novel analgesics. Here we present the selective small molecule NaV1.8 inhibitor LTGO-33 that interdicts a distinct site in a voltage-sensor domain to inhibit channel opening. These results inform the development of new analgesics for pain disorders.


Subject(s)
Voltage-Gated Sodium Channels , Humans , Animals , Dogs , Pain/drug therapy , Analgesics/pharmacology , Neurons , Action Potentials , Ganglia, Spinal , NAV1.7 Voltage-Gated Sodium Channel , Sodium Channel Blockers/pharmacology
2.
Bioorg Med Chem Lett ; 73: 128892, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35850422

ABSTRACT

NaV1.7 is an actively pursued, genetically validated, target for pain. Recently reported quinolinone sulfonamide inhibitors displayed promising selectivity profiles as well as efficacy in preclinical pain models; however, concerns about off-target liabilities associated with this series resulted in an effort to reduce the lipophilicity of these compounds. Successful prosecution of this strategy was challenging due to the opposing requirement for lipophilic inhibitors for NaV1.7 potency and in vivo clearance (CL). Deconstruction of the heterocyclic core of the quinolinone series and utilization of an intramolecular hydrogen bond to mimic the requisite pharmacophore enabled the introduction of polarity without adversely impacting CL. Ultimately, this strategy led to the identification of compound 29, which demonstrated favorable ADME and was efficacious in pre-clinical models of pain.


Subject(s)
NAV1.7 Voltage-Gated Sodium Channel , Quinolones , Humans , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Pain/drug therapy , Structure-Activity Relationship , Sulfanilamide , Sulfonamides/chemistry , Sulfonamides/pharmacology , Urea/pharmacology , Voltage-Gated Sodium Channel Blockers/chemistry
3.
SLAS Discov ; 25(5): 434-446, 2020 06.
Article in English | MEDLINE | ID: mdl-32292096

ABSTRACT

The voltage-gated sodium channel Nav1.7 is a genetically validated target for pain; pharmacological blockers are promising as a new class of nonaddictive therapeutics. The search for Nav1.7 subtype selective inhibitors requires a reliable, scalable, and sensitive assay. Previously, we developed an all-optical electrophysiology (Optopatch) Spiking HEK platform to study activity-dependent modulation of Nav1.7 in a format compatible with high-throughput screening. In this study, we benchmarked the Optopatch Spiking HEK assay with an existing validated automated electrophysiology assay on the IonWorks Barracuda (IWB) platform. In a pilot screen of 3520 compounds, which included compound plates from a random library as well as compound plates enriched for Nav1.7 inhibitors, the Optopatch Spiking HEK assay identified 174 hits, of which 143 were confirmed by IWB. The Optopatch Spiking HEK assay maintained the high reliability afforded by traditional fluorescent assays and further demonstrated comparable sensitivity to IWB measurements. We speculate that the Optopatch assay could provide an affordable high-throughput screening platform to identify novel Nav1.7 subtype selective inhibitors with diverse mechanisms of action, if coupled with a multiwell parallel optogenetic recording instrument.


Subject(s)
High-Throughput Screening Assays , NAV1.7 Voltage-Gated Sodium Channel/drug effects , Patch-Clamp Techniques , Voltage-Gated Sodium Channel Blockers/isolation & purification , Animals , CHO Cells , Cricetulus , Electrophysiological Phenomena , Electrophysiology , HEK293 Cells , Humans , NAV1.7 Voltage-Gated Sodium Channel/genetics
4.
Pain ; 161(7): 1670-1681, 2020 07.
Article in English | MEDLINE | ID: mdl-32142016

ABSTRACT

Pituitary adenylate cyclase activating polypeptide-38 (PACAP38) may play an important role in primary headaches. Preclinical evidence suggests that PACAP38 modulates trigeminal nociceptive activity mainly through PAC1 receptors while clinical studies report that plasma concentrations of PACAP38 are elevated in spontaneous attacks of cluster headache and migraine and normalize after treatment with sumatriptan. Intravenous infusion of PACAP38 induces migraine-like attacks in migraineurs and cluster-like attacks in cluster headache patients. A rodent-specific PAC1 receptor antibody Ab181 was developed, and its effect on nociceptive neuronal activity in the trigeminocervical complex was investigated in vivo in an electrophysiological model relevant to primary headaches. Ab181 is potent and selective at the rat PAC1 receptor and provides near-maximum target coverage at 10 mg/kg for more than 48 hours. Without affecting spontaneous neuronal activity, Ab181 effectively inhibits stimulus-evoked activity in the trigeminocervical complex. Immunohistochemical analysis revealed its binding in the trigeminal ganglion and sphenopalatine ganglion but not within the central nervous system suggesting a peripheral site of action. The pharmacological approach using a specific PAC1 receptor antibody could provide a novel mechanism with a potential clinical efficacy in the treatment of primary headaches.


Subject(s)
Migraine Disorders , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Animals , Headache/chemically induced , Headache/drug therapy , Humans , Nociception , Pituitary Adenylate Cyclase-Activating Polypeptide/therapeutic use , Rats
5.
Drug Metab Dispos ; 47(10): 1111-1121, 2019 10.
Article in English | MEDLINE | ID: mdl-31387871

ABSTRACT

The identification of nonopioid alternatives to treat chronic pain has received a great deal of interest in recent years. Recently, the engineering of a series of Nav1.7 inhibitory peptide-antibody conjugates has been reported, and herein, the preclinical efforts to identify novel approaches to characterize the pharmacokinetic properties of the peptide conjugates are described. A cryopreserved plated mouse hepatocyte assay was designed to measure the depletion of the peptide-antibody conjugates from the media, with a correlation being observed between percentage remaining in the media and in vivo clearance (Pearson r = -0.5525). Physicochemical (charge and hydrophobicity), receptor-binding [neonatal Fc receptor (FcRn)], and in vivo pharmacokinetic data were generated and compared with the results from our in vitro hepatocyte assay, which was hypothesized to encompass all of the aforementioned properties. Correlations were observed among hydrophobicity; FcRn binding; depletion rates from the hepatocyte assay; and ultimately, in vivo clearance. Subsequent studies identified potential roles for the low-density lipoprotein and mannose/galactose receptors in the association of the Nav1.7 peptide conjugates with mouse hepatocytes, although in vivo studies suggested that FcRn was still the primary receptor involved in determining the pharmacokinetics of the peptide conjugates. Ultimately, the use of the cryopreserved hepatocyte assay along with FcRn binding and hydrophobic interaction chromatography provided an efficient and integrated approach to rapidly triage molecules for advancement while reducing the number of in vivo pharmacokinetic studies. SIGNIFICANCE STATEMENT: Although multiple in vitro and in silico tools are available in small-molecule drug discovery, pharmacokinetic characterization of protein therapeutics is still highly dependent upon the use of in vivo studies in preclinical species. The current work demonstrates the combined use of cryopreserved hepatocytes, hydrophobic interaction chromatography, and neonatal Fc receptor binding to characterize a series of Nav1.7 peptide-antibody conjugates prior to conducting in vivo studies, thus providing a means to rapidly evaluate novel protein therapeutic platforms while concomitantly reducing the number of in vivo studies conducted in preclinical species.


Subject(s)
Chronic Pain/drug therapy , Histocompatibility Antigens Class I/metabolism , Immunoconjugates/pharmacokinetics , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Receptors, Fc/metabolism , Voltage-Gated Sodium Channel Blockers/pharmacokinetics , Administration, Intravenous , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacokinetics , Cryopreservation , Drug Evaluation, Preclinical/methods , Hepatocytes , Histocompatibility Antigens Class I/genetics , Immunoconjugates/administration & dosage , Macaca fascicularis , Male , Metabolic Clearance Rate , Mice , Mice, Knockout , Peptides/administration & dosage , Peptides/pharmacokinetics , Receptors, Fc/genetics , Tissue Distribution , Voltage-Gated Sodium Channel Blockers/administration & dosage
6.
ACS Chem Biol ; 14(4): 806-818, 2019 04 19.
Article in English | MEDLINE | ID: mdl-30875193

ABSTRACT

Drug discovery research on new pain targets with human genetic validation, including the voltage-gated sodium channel NaV1.7, is being pursued to address the unmet medical need with respect to chronic pain and the rising opioid epidemic. As part of early research efforts on this front, we have previously developed NaV1.7 inhibitory peptide-antibody conjugates with tarantula venom-derived GpTx-1 toxin peptides with an extended half-life (80 h) in rodents but only moderate in vitro activity (hNaV1.7 IC50 = 250 nM) and without in vivo activity. We identified the more potent peptide JzTx-V from our natural peptide collection and improved its selectivity against other sodium channel isoforms through positional analogueing. Here we report utilization of the JzTx-V scaffold in a peptide-antibody conjugate and architectural variations in the linker, peptide loading, and antibody attachment site. We found conjugates with 100-fold improved in vitro potency relative to those of complementary GpTx-1 analogues, but pharmacokinetic and bioimaging analyses of these JzTx-V conjugates revealed a shorter than expected plasma half-life in vivo with accumulation in the liver. In an attempt to increase circulatory serum levels, we sought the reduction of the net +6 charge of the JzTx-V scaffold while retaining a desirable NaV in vitro activity profile. The conjugate of a JzTx-V peptide analogue with a +2 formal charge maintained NaV1.7 potency with 18-fold improved plasma exposure in rodents. Balancing the loss of peptide and conjugate potency associated with the reduction of net charge necessary for improved target exposure resulted in a compound with moderate activity in a NaV1.7-dependent pharmacodynamic model but requires further optimization to identify a conjugate that can fully engage NaV1.7 in vivo.


Subject(s)
Immunoconjugates , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Peptides/chemistry , Spider Venoms/chemistry , Voltage-Gated Sodium Channel Blockers , Animals , Antibodies/chemistry , Drug Discovery , Humans , Immunoconjugates/chemistry , Immunoconjugates/pharmacokinetics , Male , Mice , Molecular Targeted Therapy , NAV1.7 Voltage-Gated Sodium Channel/immunology , Peptides/pharmacokinetics , Spider Venoms/pharmacokinetics , Voltage-Gated Sodium Channel Blockers/chemistry , Voltage-Gated Sodium Channel Blockers/pharmacokinetics
7.
J Med Chem ; 61(21): 9500-9512, 2018 11 08.
Article in English | MEDLINE | ID: mdl-30346167

ABSTRACT

Inhibitors of the voltage-gated sodium channel NaV1.7 are being investigated as pain therapeutics due to compelling human genetics. We previously identified NaV1.7-inhibitory peptides GpTx-1 and JzTx-V from tarantula venom screens. Potency and selectivity were modulated through attribute-based positional scans of native residues via chemical synthesis. Herein, we report JzTx-V lead optimization to identify a pharmacodynamically active peptide variant. Molecular docking of peptide ensembles from NMR into a homology model-derived NaV1.7 structure supported prioritization of key residues clustered on a hydrophobic face of the disulfide-rich folded peptide for derivatization. Replacing Trp24 with 5-Br-Trp24 identified lead peptides with activity in electrophysiology assays in engineered and neuronal cells. 5-Br-Trp24 containing peptide AM-6120 was characterized in X-ray crystallography and pharmacokinetic studies and blocked histamine-induced pruritis in mice after subcutaneous administration, demonstrating systemic NaV1.7-dependent pharmacodynamics. Our data suggests a need for high target coverage based on plasma exposure for impacting in vivo end points with selectivity-optimized peptidic NaV1.7 inhibitors.


Subject(s)
Drug Discovery , Histamine/adverse effects , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Peptides/chemistry , Peptides/pharmacology , Pruritus/drug therapy , Spider Venoms/chemistry , Animals , HEK293 Cells , Humans , Mice , Molecular Docking Simulation , NAV1.7 Voltage-Gated Sodium Channel/chemistry , Peptides/pharmacokinetics , Peptides/therapeutic use , Protein Conformation , Protein Folding , Pruritus/chemically induced , Structure-Activity Relationship , Tissue Distribution , Voltage-Gated Sodium Channel Blockers/chemistry , Voltage-Gated Sodium Channel Blockers/pharmacokinetics , Voltage-Gated Sodium Channel Blockers/pharmacology , Voltage-Gated Sodium Channel Blockers/therapeutic use
8.
PLoS One ; 13(5): e0196791, 2018.
Article in English | MEDLINE | ID: mdl-29723257

ABSTRACT

Identification of voltage-gated sodium channel NaV1.7 inhibitors for chronic pain therapeutic development is an area of vigorous pursuit. In an effort to identify more potent leads compared to our previously reported GpTx-1 peptide series, electrophysiology screening of fractionated tarantula venom discovered the NaV1.7 inhibitory peptide JzTx-V from the Chinese earth tiger tarantula Chilobrachys jingzhao. The parent peptide displayed nominal selectivity over the skeletal muscle NaV1.4 channel. Attribute-based positional scan analoging identified a key Ile28Glu mutation that improved NaV1.4 selectivity over 100-fold, and further optimization yielded the potent and selective peptide leads AM-8145 and AM-0422. NMR analyses revealed that the Ile28Glu substitution changed peptide conformation, pointing to a structural rationale for the selectivity gains. AM-8145 and AM-0422 as well as GpTx-1 and HwTx-IV competed for ProTx-II binding in HEK293 cells expressing human NaV1.7, suggesting that these NaV1.7 inhibitory peptides interact with a similar binding site. AM-8145 potently blocked native tetrodotoxin-sensitive (TTX-S) channels in mouse dorsal root ganglia (DRG) neurons, exhibited 30- to 120-fold selectivity over other human TTX-S channels and exhibited over 1,000-fold selectivity over other human tetrodotoxin-resistant (TTX-R) channels. Leveraging NaV1.7-NaV1.5 chimeras containing various voltage-sensor and pore regions, AM-8145 mapped to the second voltage-sensor domain of NaV1.7. AM-0422, but not the inactive peptide analog AM-8374, dose-dependently blocked capsaicin-induced DRG neuron action potential firing using a multi-electrode array readout and mechanically-induced C-fiber spiking in a saphenous skin-nerve preparation. Collectively, AM-8145 and AM-0422 represent potent, new engineered NaV1.7 inhibitory peptides derived from the JzTx-V scaffold with improved NaV selectivity and biological activity in blocking action potential firing in both DRG neurons and C-fibers.


Subject(s)
Analgesics/isolation & purification , NAV1.7 Voltage-Gated Sodium Channel/drug effects , Peptides/chemistry , Sodium Channel Blockers/isolation & purification , Spider Venoms/chemistry , Action Potentials/drug effects , Amino Acid Substitution , Analgesics/pharmacology , Animals , Capsaicin/pharmacology , Cell Line , Drug Evaluation, Preclinical , Ganglia, Spinal/drug effects , Humans , Male , Mice, Inbred C57BL , Mutagenesis, Site-Directed , Nerve Fibers, Unmyelinated/drug effects , Nuclear Magnetic Resonance, Biomolecular , Patch-Clamp Techniques , Physical Stimulation , Protein Engineering , Recombinant Proteins/drug effects , Sodium Channel Blockers/pharmacology , Structure-Activity Relationship , Tetrodotoxin/pharmacology
9.
Bioorg Med Chem Lett ; 28(11): 2103-2108, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29709252

ABSTRACT

Recently, the identification of several classes of aryl sulfonamides and acyl sulfonamides that potently inhibit NaV1.7 and demonstrate high levels of selectivity over other NaV isoforms have been reported. The fully ionizable nature of these inhibitors has been shown to be an important part of the pharmacophore for the observed potency and isoform selectivity. The requirement of this functionality, however, has presented challenges associated with optimization toward inhibitors with drug-like properties and minimal off-target activity. In an effort to obviate these challenges, we set out to develop an orally bioavailable, selective NaV1.7 inhibitor, lacking these acidic functional groups. Herein, we report the discovery of a novel series of inhibitors wherein a triazolesulfone has been designed to serve as a bioisostere for the acyl sulfonamide. This work culminated in the delivery of a potent series of inhibitors which demonstrated good levels of selectivity over NaV1.5 and favorable pharmacokinetics in rodents.


Subject(s)
NAV1.7 Voltage-Gated Sodium Channel/metabolism , Sulfonamides/pharmacology , Animals , Dose-Response Relationship, Drug , Humans , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Molecular Structure , Rats , Structure-Activity Relationship , Sulfonamides/chemistry
10.
ACS Chem Biol ; 12(9): 2427-2435, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28800217

ABSTRACT

The voltage-gated sodium channel NaV1.7 is a genetically validated pain target under investigation for the development of analgesics. A therapeutic with a less frequent dosing regimen would be of value for treating chronic pain; however functional NaV1.7 targeting antibodies are not known. In this report, we describe NaV1.7 inhibitory peptide-antibody conjugates as an alternate construct for potential prolonged channel blockade through chemical derivatization of engineered antibodies. We previously identified NaV1.7 inhibitory peptide GpTx-1 from tarantula venom and optimized its potency and selectivity. Tethering GpTx-1 peptides to antibodies bifunctionally couples FcRn-based antibody recycling attributes to the NaV1.7 targeting function of the peptide warhead. Herein, we conjugated a GpTx-1 peptide to specific engineered cysteines in a carrier anti-2,4-dinitrophenol monoclonal antibody using polyethylene glycol linkers. The reactivity of 13 potential cysteine conjugation sites in the antibody scaffold was tuned using a model alkylating agent. Subsequent reactions with the peptide identified cysteine locations with the highest conversion to desired conjugates, which blocked NaV1.7 currents in whole cell electrophysiology. Variations in attachment site, linker, and peptide loading established design parameters for potency optimization. Antibody conjugation led to in vivo half-life extension by 130-fold relative to a nonconjugated GpTx-1 peptide and differential biodistribution to nerve fibers in wild-type but not NaV1.7 knockout mice. This study describes the optimization and application of antibody derivatization technology to functionally inhibit NaV1.7 in engineered and neuronal cells.


Subject(s)
Immunoconjugates/pharmacology , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Peptides/pharmacology , Voltage-Gated Sodium Channel Blockers/pharmacology , Animals , HEK293 Cells , Humans , Immunoconjugates/chemistry , Immunoconjugates/pharmacokinetics , Male , Mice , Models, Molecular , Peptides/chemistry , Peptides/pharmacokinetics , Tissue Distribution , Voltage-Gated Sodium Channel Blockers/chemistry , Voltage-Gated Sodium Channel Blockers/pharmacokinetics
11.
Bioorg Med Chem Lett ; 27(16): 3817-3824, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28684121

ABSTRACT

The NaV1.7 ion channel has garnered considerable attention as a target for the treatment of pain. Herein we detail the discovery and structure-activity relationships of a novel series of biaryl amides. Optimization led to the identification of several state-dependent, potent and metabolically stable inhibitors which demonstrated promising levels of selectivity over NaV1.5 and good rat pharmacokinetics. Compound 18, which demonstrated preferential inhibition of a slow inactivated state of NaV1.7, was advanced into a rat formalin study where upon reaching unbound drug levels several fold over the rat NaV1.7 IC50 it failed to demonstrate a robust reduction in nociceptive behavior.


Subject(s)
Amides/pharmacology , Drug Discovery , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Amides/chemical synthesis , Amides/chemistry , Animals , Dose-Response Relationship, Drug , Humans , Mice , Molecular Structure , Rats , Structure-Activity Relationship
12.
Bioorg Med Chem Lett ; 27(15): 3477-3485, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28629594

ABSTRACT

The voltage-gated sodium channel NaV1.7 has received much attention from the scientific community due to compelling human genetic data linking gain- and loss-of-function mutations to pain phenotypes. Despite this genetic validation of NaV1.7 as a target for pain, high quality pharmacological tools facilitate further understanding of target biology, establishment of target coverage requirements and subsequent progression into the clinic. Within the sulfonamide class of inhibitors, reduced potency on rat NaV1.7 versus human NaV1.7 was observed, rendering in vivo rat pharmacology studies challenging. Herein, we report the discovery and optimization of novel benzoxazine sulfonamide inhibitors of human, rat and mouse NaV1.7 which enabled pharmacological assessment in traditional behavioral rodent models of pain and in turn, established a connection between formalin-induced pain and histamine-induced pruritus in mice. The latter represents a simple and efficient means of measuring target engagement.


Subject(s)
Benzoxazines/chemistry , Benzoxazines/pharmacology , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Sulfonamides/chemistry , Sulfonamides/pharmacology , Voltage-Gated Sodium Channel Blockers/chemistry , Voltage-Gated Sodium Channel Blockers/pharmacology , Analgesics/chemistry , Analgesics/pharmacokinetics , Analgesics/pharmacology , Analgesics/therapeutic use , Animals , Benzoxazines/pharmacokinetics , Benzoxazines/therapeutic use , Humans , Male , Mice , Mice, Inbred C57BL , Molecular Docking Simulation , Pain/drug therapy , Pain/metabolism , Rats , Rats, Sprague-Dawley , Sulfonamides/pharmacokinetics , Sulfonamides/therapeutic use , Voltage-Gated Sodium Channel Blockers/pharmacokinetics , Voltage-Gated Sodium Channel Blockers/therapeutic use
13.
J Pharmacol Exp Ther ; 362(1): 146-160, 2017 07.
Article in English | MEDLINE | ID: mdl-28473457

ABSTRACT

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.


Subject(s)
NAV1.7 Voltage-Gated Sodium Channel/drug effects , Sodium Channel Blockers/pharmacology , Action Potentials/drug effects , Animals , Behavior, Animal/drug effects , Dose-Response Relationship, Drug , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Humans , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Myocardium/metabolism , Neurons/drug effects , Pain/prevention & control , Pain/psychology , Patch-Clamp Techniques , Pruritus/prevention & control , Pruritus/psychology , Quinolones/pharmacology , Small Molecule Libraries , Stereoisomerism , Sulfonamides/pharmacology
15.
J Med Chem ; 60(14): 5990-6017, 2017 07 27.
Article in English | MEDLINE | ID: mdl-28324649

ABSTRACT

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.


Subject(s)
NAV1.7 Voltage-Gated Sodium Channel/metabolism , Quinolones/chemistry , Sulfonamides/chemistry , Voltage-Gated Sodium Channel Blockers/chemistry , Analgesics/chemistry , Analgesics/pharmacokinetics , Analgesics/pharmacology , Animals , Capsaicin , Cell Line , Dogs , Histamine , Mice, Inbred C57BL , Molecular Docking Simulation , Pain/chemically induced , Pain/prevention & control , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Pruritus/chemically induced , Pruritus/prevention & control , Quinolones/administration & dosage , Quinolones/chemical synthesis , Quinolones/pharmacokinetics , Quinolones/pharmacology , Rats , Structure-Activity Relationship , Sulfonamides/administration & dosage , Sulfonamides/chemical synthesis , Sulfonamides/pharmacokinetics , Sulfonamides/pharmacology , Voltage-Gated Sodium Channel Blockers/pharmacokinetics , Voltage-Gated Sodium Channel Blockers/pharmacology
16.
J Med Chem ; 60(14): 5969-5989, 2017 07 27.
Article in English | MEDLINE | ID: mdl-28287723

ABSTRACT

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.


Subject(s)
Isoquinolines/chemistry , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Sulfonamides/chemistry , Voltage-Gated Sodium Channel Blockers/chemistry , Animals , Cell Line , Cytochrome P-450 CYP3A/biosynthesis , Cytochrome P-450 CYP3A Inhibitors/chemistry , Cytochrome P-450 CYP3A Inhibitors/pharmacokinetics , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Dogs , Enzyme Induction , Histamine , Humans , Isoquinolines/administration & dosage , Isoquinolines/pharmacokinetics , Male , Mice, Inbred C57BL , Pregnane X Receptor , Pruritus/chemically induced , Pruritus/prevention & control , Rats , Receptors, Steroid/agonists , Structure-Activity Relationship , Sulfonamides/administration & dosage , Sulfonamides/pharmacokinetics , Sulfonamides/pharmacology , Voltage-Gated Sodium Channel Blockers/pharmacokinetics , Voltage-Gated Sodium Channel Blockers/pharmacology
17.
ACS Med Chem Lett ; 7(12): 1062-1067, 2016 Dec 08.
Article in English | MEDLINE | ID: mdl-27994738

ABSTRACT

Human genetic evidence has identified the voltage-gated sodium channel NaV1.7 as an attractive target for the treatment of pain. We initially identified naphthalene sulfonamide 3 as a potent and selective inhibitor of NaV1.7. Optimization to reduce biliary clearance by balancing hydrophilicity and hydrophobicity (Log D) while maintaining NaV1.7 potency led to the identification of quinazoline 16 (AM-2099). Compound 16 demonstrated a favorable pharmacokinetic profile in rat and dog and demonstrated dose-dependent reduction of histamine-induced scratching bouts in a mouse behavioral model following oral dosing.

18.
F1000Res ; 5: 2764, 2016.
Article in English | MEDLINE | ID: mdl-27990272

ABSTRACT

Identification of small and large molecule pain therapeutics that target the genetically validated voltage-gated sodium channel Na V1.7 is a challenging endeavor under vigorous pursuit. The monoclonal antibody SVmab1 was recently published to bind the Na V1.7 DII voltage sensor domain and block human Na V1.7 sodium currents in heterologous cells. We produced purified SVmab1 protein based on publically available sequence information, and evaluated its activity in a battery of binding and functional assays. Herein, we report that our recombinant SVmAb1 does not bind peptide immunogen or purified Na V1.7 DII voltage sensor domain via ELISA, and does not bind Na V1.7 in live HEK293, U-2 OS, and CHO-K1 cells via FACS. Whole cell manual patch clamp electrophysiology protocols interrogating diverse Na V1.7 gating states in HEK293 cells, revealed that recombinant SVmab1 does not block Na V1.7 currents to an extent greater than observed with an isotype matched control antibody. Collectively, our results show that recombinant SVmab1 monoclonal antibody does not bind Na V1.7 target sequences or specifically inhibit Na V1.7 current.

19.
Mol Pain ; 122016.
Article in English | MEDLINE | ID: mdl-27899696

ABSTRACT

The transient receptor potential ankyrin 1 (TRPA1) channel has been implicated in pathophysiological processes that include asthma, cough, and inflammatory pain. Agonists of TRPA1 such as mustard oil and its key component allyl isothiocyanate (AITC) cause pain and neurogenic inflammation in humans and rodents, and TRPA1 antagonists have been reported to be effective in rodent models of pain. In our pursuit of TRPA1 antagonists as potential therapeutics, we generated AMG0902, a potent (IC90 of 300 nM against rat TRPA1), selective, brain penetrant (brain to plasma ratio of 0.2), and orally bioavailable small molecule TRPA1 antagonist. AMG0902 reduced mechanically evoked C-fiber action potential firing in a skin-nerve preparation from mice previously injected with complete Freund's adjuvant, supporting the role of TRPA1 in inflammatory mechanosensation. In vivo target coverage of TRPA1 by AMG0902 was demonstrated by the prevention of AITC-induced flinching/licking in rats. However, oral administration of AMG0902 to rats resulted in little to no efficacy in models of inflammatory, mechanically evoked hypersensitivity; and no efficacy was observed in a neuropathic pain model. Unbound plasma concentrations achieved in pain models were about 4-fold higher than the IC90 concentration in the AITC target coverage model, suggesting that either greater target coverage is required for efficacy in the pain models studied or TRPA1 may not contribute significantly to the underlying mechanisms.


Subject(s)
Hyperalgesia/metabolism , Inflammation/complications , Sciatica/complications , TRPC Cation Channels/metabolism , Action Potentials/drug effects , Action Potentials/genetics , Amines/therapeutic use , Analgesics/therapeutic use , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , CHO Cells , Cricetulus , Cyclohexanecarboxylic Acids/therapeutic use , Exploratory Behavior/drug effects , Freund's Adjuvant/toxicity , Gabapentin , Hyperalgesia/drug therapy , Inflammation/chemically induced , Inflammation/drug therapy , Male , Mice, Inbred C57BL , Mice, Knockout , Naproxen/pharmacology , Nerve Fibers, Unmyelinated/drug effects , Nerve Fibers, Unmyelinated/physiology , Pain Threshold/drug effects , Rats , Rats, Sprague-Dawley , Sciatica/drug therapy , TRPA1 Cation Channel , TRPC Cation Channels/antagonists & inhibitors , TRPC Cation Channels/genetics , gamma-Aminobutyric Acid/therapeutic use
20.
J Med Chem ; 59(17): 7818-39, 2016 09 08.
Article in English | MEDLINE | ID: mdl-27441383

ABSTRACT

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.


Subject(s)
Benzamides/chemistry , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Sulfonamides/chemistry , Voltage-Gated Sodium Channel Blockers/chemistry , Animals , Benzamides/chemical synthesis , Benzamides/pharmacokinetics , Benzamides/pharmacology , Cell Line , Female , Histamine , Humans , Male , Mice, Inbred C57BL , Microsomes, Liver/metabolism , Molecular Docking Simulation , Pruritus/chemically induced , Pruritus/drug therapy , Radioligand Assay , Rats , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/pharmacokinetics , Sulfonamides/pharmacology , Voltage-Gated Sodium Channel Blockers/chemical synthesis , Voltage-Gated Sodium Channel Blockers/pharmacokinetics , Voltage-Gated Sodium Channel Blockers/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...