1
|
Alsaloum M, Dib-Hajj SD, Page DA, Ruben PC, Krainer AR, Waxman SG. Voltage-gated sodium channels in excitable cells as drug targets. Nat Rev Drug Discov 2025:10.1038/s41573-024-01108-x. [PMID: 39901031 DOI: 10.1038/s41573-024-01108-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2024] [Indexed: 02/05/2025]
Abstract
Excitable cells - including neurons, muscle cells and cardiac myocytes - are unique in expressing high densities of voltage-gated sodium (NaV) channels. This molecular adaptation enables these cells to produce action potentials, and is essential to their function. With the advent of the molecular revolution, the concept of 'the' sodium channel has been supplanted by understanding that excitable cells in mammals can express any of nine different forms of sodium channels (NaV1.1-NaV1.9). Selective expression in particular types of cells, together with a key role in controlling action potential firing, makes some of these NaV subtypes especially attractive molecular targets for drug development. Although these different channel subtypes display a common overall structure, differences in their amino acid sequences have provided a basis for the development of subtype-specific drugs. This approach has resulted in exciting progress in the development of drugs for epilepsy, cardiac disorders and pain. In this Review, we discuss recent progress in the development of drugs that selectively target each of the sodium channel subtypes.
Collapse
Affiliation(s)
- Matthew Alsaloum
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Dana A Page
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Peter C Ruben
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | | | - Stephen G Waxman
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
2
|
Sun S, Chowdhury S, Hemeon I, Hasan A, Wilson MS, Bergeron P, Jia Q, Zenova AY, Grimwood ME, Gong W, Decker SM, Bichler P, Andrez JC, Focken T, Ngyuen T, Zhu J, White AD, Bankar G, Howard S, Chang E, Khakh K, Lin S, Dean R, Johnson JP, Chang JH, Hackos DH, McKerrall SJ, Sellers B, Ortwine DF, Cohen CJ, Safina BS, Sutherlin DP, Dehnhardt CM. Discovery of novel cyclopentane carboxylic acids as potent and selective inhibitors of Na V1.7. Bioorg Med Chem Lett 2025; 116:130033. [PMID: 39580005 DOI: 10.1016/j.bmcl.2024.130033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/28/2024] [Accepted: 11/17/2024] [Indexed: 11/25/2024]
Abstract
Discovery efforts leading to the identification of cyclopentane carboxylic acid 31, a potent inhibitor of NaV1.7 that showed high selectivity over NaV1.5 and exhibited robust analgesic effects in an inherited erythromelalgia (IEM) transgenic mouse assay, are described herein. Key design elements that culminated in the discovery of 31 include exploration of proline substituents, replacement of the proline warhead with cyclopentane carboxylic acid, that led to significantly boosted NaV1.7 potency, and replacement of the metabolically labile adamantane motif with the 2,6-dichlorobenzyl substituted piperidine system, that addressed metabolic instability and led to a significant improvement in PK.
Collapse
Affiliation(s)
- Shaoyi Sun
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada.
| | - Sultan Chowdhury
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Ivan Hemeon
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Abid Hasan
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Michael S Wilson
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | | | - Qi Jia
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Alla Y Zenova
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Mike E Grimwood
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Wei Gong
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Shannon M Decker
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Paul Bichler
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | | | - Thilo Focken
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Theresa Ngyuen
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080-4990, USA
| | - Jiuxiang Zhu
- Chempartner, Building No. 5, 998 Halei Rd., Zhangjiang Hi-Tech Park, Pudong New Area, Shanghai, PR China
| | - Andrew D White
- Chempartner, Building No. 5, 998 Halei Rd., Zhangjiang Hi-Tech Park, Pudong New Area, Shanghai, PR China
| | - Girish Bankar
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Sarah Howard
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080-4990, USA
| | - Elaine Chang
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Kuldip Khakh
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Sophia Lin
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Richard Dean
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - J P Johnson
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Jae H Chang
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080-4990, USA
| | - David H Hackos
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080-4990, USA
| | | | - Ben Sellers
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080-4990, USA
| | - Dan F Ortwine
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080-4990, USA
| | - Charles J Cohen
- Xenon Pharmaceuticals Inc., 3650 Gilmore Way, Burnaby, BC V5G 4W8, Canada
| | - Brian S Safina
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080-4990, USA
| | | | | |
Collapse
|
3
|
Lopez-Mateos D, Harris BJ, Hernández-González A, Narang K, Yarov-Yarovoy V. Harnessing Deep Learning Methods for Voltage-Gated Ion Channel Drug Discovery. Physiology (Bethesda) 2025; 40:0. [PMID: 39189871 DOI: 10.1152/physiol.00029.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/16/2024] [Accepted: 08/18/2024] [Indexed: 08/28/2024] Open
Abstract
Voltage-gated ion channels (VGICs) are pivotal in regulating electrical activity in excitable cells and are critical pharmaceutical targets for treating many diseases including cardiac arrhythmia and neuropathic pain. Despite their significance, challenges such as achieving target selectivity persist in VGIC drug development. Recent progress in deep learning, particularly diffusion models, has enabled the computational design of protein binders for any clinically relevant protein based solely on its structure. These developments coincide with a surge in experimental structural data for VGICs, providing a rich foundation for computational design efforts. This review explores the recent advancements in computational protein design using deep learning and diffusion methods, focusing on their application in designing protein binders to modulate VGIC activity. We discuss the potential use of these methods to computationally design protein binders targeting different regions of VGICs, including the pore domain, voltage-sensing domains, and interface with auxiliary subunits. We provide a comprehensive overview of the different design scenarios, discuss key structural considerations, and address the practical challenges in developing VGIC-targeting protein binders. By exploring these innovative computational methods, we aim to provide a framework for developing novel strategies that could significantly advance VGIC pharmacology and lead to the discovery of effective and safe therapeutics.
Collapse
Affiliation(s)
- Diego Lopez-Mateos
- Department of Physiology and Membrane Biology, University of California School of Medicine, Davis, California, United States
- Biophysics Graduate Group, University of California School of Medicine, Davis, California, United States
| | - Brandon John Harris
- Department of Physiology and Membrane Biology, University of California School of Medicine, Davis, California, United States
- Biophysics Graduate Group, University of California School of Medicine, Davis, California, United States
| | - Adriana Hernández-González
- Department of Physiology and Membrane Biology, University of California School of Medicine, Davis, California, United States
- Biophysics Graduate Group, University of California School of Medicine, Davis, California, United States
| | - Kush Narang
- Department of Physiology and Membrane Biology, University of California School of Medicine, Davis, California, United States
| | - Vladimir Yarov-Yarovoy
- Department of Physiology and Membrane Biology, University of California School of Medicine, Davis, California, United States
- Biophysics Graduate Group, University of California School of Medicine, Davis, California, United States
- Department of Anesthesiology and Pain Medicine, University of California School of Medicine, Davis, California, United States
| |
Collapse
|
4
|
Hestehave S, Allen HN, Gomez K, Duran P, Calderon-Rivera A, Loya-López S, Rodríguez-Palma EJ, Khanna R. Small molecule targeting Na V 1.7 via inhibition of CRMP2-Ubc9 interaction reduces pain-related outcomes in a rodent osteoarthritic model. Pain 2025; 166:99-111. [PMID: 39106443 PMCID: PMC11649477 DOI: 10.1097/j.pain.0000000000003357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 05/30/2024] [Indexed: 08/09/2024]
Abstract
ABSTRACT Osteoarthritis (OA) is a highly prevalent and disabling joint disease, characterized by pathological progressive joint deformation and clinical symptoms of pain. Disease-modifying treatments remain unavailable, and pain-mitigation is often suboptimal, but recent studies suggest beneficial effects by inhibition of the voltage-gated sodium channel Na V 1.7. We previously identified compound 194 as an indirect inhibitor of Na V 1.7 by preventing SUMOylation of the Na V 1.7-trafficking protein, collapsin response mediator protein 2. Compound 194 reduces the functional activity of Na V 1.7 channels and produces effective analgesia in a variety of acute and neuropathic pain models. However, its effectiveness has not yet been evaluated in models of OA. Here, we explore the effects of 194 on pain-related outcomes in the OA-like monoiodoacetate model using behavioral assessment, biochemistry, novel in vivo fiber photometry, and patch clamp electrophysiology. We found that the monoiodoacetate model induced (1) increased pain-like behaviors and calcium responses of glutamatergic neurons in the parabrachial nucleus after evoked cold and mechanical stimuli, (2) conditioned place aversion to mechanical stimulation, (3) functional weight bearing asymmetry, (4) increased sodium currents in dorsal root ganglia neurons, and (5) increased calcitonin gene-related peptide-release in the spinal cord. Crucially, administration of 194 improved all these pain-related outcomes. Collectively, these findings support indirect inhibition of Na V 1.7 as an effective treatment of OA-related pain through the inhibition of collapsin response mediator protein 2-SUMOylation via compound 194.
Collapse
Affiliation(s)
- Sara Hestehave
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York 10010, USA
- Pain Research Center, New York University, New York, NY 10010, USA
| | - Heather N. Allen
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York 10010, USA
- Pain Research Center, New York University, New York, NY 10010, USA
| | - Kimberly Gomez
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York 10010, USA
- Pain Research Center, New York University, New York, NY 10010, USA
| | - Paz Duran
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York 10010, USA
- Pain Research Center, New York University, New York, NY 10010, USA
| | - Aida Calderon-Rivera
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York 10010, USA
- Pain Research Center, New York University, New York, NY 10010, USA
| | - Santiago Loya-López
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York 10010, USA
- Pain Research Center, New York University, New York, NY 10010, USA
| | - Erick J. Rodríguez-Palma
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York 10010, USA
- Pain Research Center, New York University, New York, NY 10010, USA
| | - Rajesh Khanna
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York 10010, USA
- Pain Research Center, New York University, New York, NY 10010, USA
| |
Collapse
|
5
|
Pulskamp TG, Johnson LM, Berlau DJ. Novel non-opioid analgesics in pain management. Pain Manag 2024; 14:641-651. [PMID: 39692452 PMCID: PMC11702995 DOI: 10.1080/17581869.2024.2442292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 12/11/2024] [Indexed: 12/19/2024] Open
Abstract
Effective pain management has long been hindered by the limitations and risks associated with opioid analgesics, necessitating the exploration of novel, non-opioid alternatives. A comprehensive literature search was conducted using PubMed and Google Scholar during October and November 2024 to identify studies on emerging non-opioid pain management therapeutics. This review evaluates three promising classes of mechanism-specific therapeutics: nerve growth factor (NGF) monoclonal antibodies, transient receptor potential vanilloid 1 (TRPV1) antagonists, and selective sodium channel blockers. By targeting distinct pathways involved in pain sensation, these therapies aim to provide relief for various pain types, including chronic, inflammatory, and neuropathic pain, with potentially fewer side effects. Through a detailed analysis of their mechanisms of action and current evidence, this review highlights the clinical potential of each class, addressing both their efficacy and safety challenges. Ultimately, these emerging therapies represent significant advancements in non-opioid pain management, with the potential to reshape standard approaches to patient care.
Collapse
|
6
|
Mulcahy JV, Beckley JT, Klas SD, Odink DA, Delwig A, Pajouhesh H, Monteleone D, Zhou X, Du Bois J, Yeomans DC, Luu G, Hunter JC. ST-2560, a selective inhibitor of the Na V1.7 sodium channel, affects nocifensive and cardiovascular reflexes in non-human primates. Br J Pharmacol 2024; 181:3160-3171. [PMID: 38715413 DOI: 10.1111/bph.16398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 02/25/2024] [Accepted: 02/26/2024] [Indexed: 08/03/2024] Open
Abstract
BACKGROUND AND PURPOSE The voltage-gated sodium channel isoform NaV1.7 is a high-interest target for the development of non-opioid analgesics due to its preferential expression in pain-sensing neurons. NaV1.7 is also expressed in autonomic neurons, yet its contribution to involuntary visceral reflexes has received limited attention. The small molecule inhibitor ST-2560 was advanced into pain behaviour and cardiovascular models to understand the pharmacodynamic effects of selective inhibition of NaV1.7. EXPERIMENTAL APPROACH Potency of ST-2560 at NaV1.7 and off-target ion channels was evaluated by whole-cell patch-clamp electrophysiology. Effects on nocifensive reflexes were assessed in non-human primate (NHP) behavioural models, employing the chemical capsaicin and mechanical stimuli. Cardiovascular parameters were monitored continuously in freely-moving, telemetered NHPs following administration of vehicle and ST-2560. KEY RESULTS ST-2560 is a potent inhibitor (IC50 = 39 nM) of NaV1.7 in primates with ≥1000-fold selectivity over other isoforms of the human NaV1.x family. Following systemic administration, ST-2560 (0.1-0.3 mg·kg-1, s.c.) suppressed noxious mechanical- and chemical-evoked reflexes at free plasma concentrations threefold to fivefold above NaV1.7 IC50. ST-2560 (0.1-1.0 mg·kg-1, s.c.) also produced changes in haemodynamic parameters, most notably a 10- to 20-mmHg reduction in systolic and diastolic arterial blood pressure, at similar exposures. CONCLUSIONS AND IMPLICATIONS Acute pharmacological inhibition of NaV1.7 is antinociceptive, but also has the potential to impact the cardiovascular system. Further work is merited to understand the role of NaV1.7 in autonomic ganglia involved in the control of heart rate and blood pressure, and the effect of selective NaV1.7 inhibition on cardiovascular function.
Collapse
Affiliation(s)
- John V Mulcahy
- SiteOne Therapeutics, Inc., South San Francisco, California, USA
| | - Jacob T Beckley
- SiteOne Therapeutics, Inc., South San Francisco, California, USA
| | - Sheri D Klas
- SiteOne Therapeutics, Inc., South San Francisco, California, USA
| | - Debra A Odink
- SiteOne Therapeutics, Inc., South San Francisco, California, USA
| | - Anton Delwig
- SiteOne Therapeutics, Inc., South San Francisco, California, USA
| | - Hassan Pajouhesh
- SiteOne Therapeutics, Inc., South San Francisco, California, USA
| | | | - Xiang Zhou
- SiteOne Therapeutics, Inc., South San Francisco, California, USA
| | - Justin Du Bois
- Department of Chemistry, Stanford University, Stanford, California, USA
| | - David C Yeomans
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - George Luu
- SiteOne Therapeutics, Inc., South San Francisco, California, USA
| | - John C Hunter
- SiteOne Therapeutics, Inc., South San Francisco, California, USA
| |
Collapse
|
7
|
Le Franc A, Da Silva A, Lepetre-Mouelhi S. Nanomedicine and voltage-gated sodium channel blockers in pain management: a game changer or a lost cause? Drug Deliv Transl Res 2024; 14:2112-2145. [PMID: 38861139 DOI: 10.1007/s13346-024-01615-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2024] [Indexed: 06/12/2024]
Abstract
Pain, a complex and debilitating condition affecting millions globally, is a significant concern, especially in the context of post-operative recovery. This comprehensive review explores the complexity of pain and its global impact, emphasizing the modulation of voltage-gated sodium channels (VGSC or NaV channels) as a promising avenue for pain management with the aim of reducing reliance on opioids. The article delves into the role of specific NaV isoforms, particularly NaV 1.7, NaV 1.8, and NaV 1.9, in pain process and discusses the development of sodium channel blockers to target these isoforms precisely. Traditional local anesthetics and selective NaV isoform inhibitors, despite showing varying efficacy in pain management, face challenges in systemic distribution and potential side effects. The review highlights the potential of nanomedicine in improving the delivery of local anesthetics, toxins and selective NaV isoform inhibitors for a targeted and sustained release at the site of pain. This innovative strategy seeks to improve drug bioavailability, minimize systemic exposure, and optimize therapeutic outcomes, holding significant promise for secure pain management and enhancing the quality of life for individuals recovering from surgical procedures or suffering from chronic pain.
Collapse
Affiliation(s)
- Adélaïde Le Franc
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400, Orsay, France
| | - Alexandre Da Silva
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400, Orsay, France
| | | |
Collapse
|
8
|
Kim JS, Meeker S, Ru F, Tran M, Zabka TS, Hackos D, Undem BJ. Role of Na V1.7 in postganglionic sympathetic nerve function in human and guinea-pig arteries. J Physiol 2024; 602:3505-3518. [PMID: 38743485 PMCID: PMC11250678 DOI: 10.1113/jp286538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 04/24/2024] [Indexed: 05/16/2024] Open
Abstract
NaV1.7 plays a crucial role in inducing and conducting action potentials in pain-transducing sensory nociceptor fibres, suggesting that NaV1.7 blockers could be effective non-opioid analgesics. While SCN9A is expressed in both sensory and autonomic neurons, its functional role in the autonomic system remains less established. Our single neuron rt-PCR analysis revealed that 82% of sympathetic neurons isolated from guinea-pig stellate ganglia expressed NaV1.7 mRNA, with NaV1.3 being the only other tetrodotoxin-sensitive channel expressed in approximately 50% of neurons. We investigated the role of NaV1.7 in conducting action potentials in postganglionic sympathetic nerves and in the sympathetic adrenergic contractions of blood vessels using selective NaV1.7 inhibitors. Two highly selective NaV1.7 blockers, GNE8493 and PF 05089771, significantly inhibited postganglionic compound action potentials by approximately 70% (P < 0.01), with residual activity being blocked by the NaV1.3 inhibitor, ICA 121431. Electrical field stimulation (EFS) induced rapid contractions in guinea-pig isolated aorta, pulmonary arteries, and human isolated pulmonary arteries via stimulation of intrinsic nerves, which were inhibited by prazosin or the NaV1 blocker tetrodotoxin. Our results demonstrated that blocking NaV1.7 with GNE8493, PF 05089771, or ST2262 abolished or strongly inhibited sympathetic adrenergic responses in guinea-pigs and human vascular smooth muscle. These findings support the hypothesis that pharmacologically inhibiting NaV1.7 could potentially reduce sympathetic and parasympathetic function in specific vascular beds and airways. KEY POINTS: 82% of sympathetic neurons isolated from the stellate ganglion predominantly express NaV1.7 mRNA. NaV1.7 blockers inhibit action potential conduction in postganglionic sympathetic nerves. NaV1.7 blockade substantially inhibits sympathetic nerve-mediated adrenergic contractions in human and guinea-pig blood vessels. Pharmacologically blocking NaV1.7 profoundly affects sympathetic and parasympathetic responses in addition to sensory fibres, prompting exploration into the broader physiological consequences of NaV1.7 mutations on autonomic nerve activity.
Collapse
Affiliation(s)
- Joyce S Kim
- Johns Hopkins School of Medicine, Division of Clinical Immunology, Baltimore, MD, USA
| | - Sonya Meeker
- Johns Hopkins School of Medicine, Division of Clinical Immunology, Baltimore, MD, USA
| | - Fei Ru
- Johns Hopkins School of Medicine, Division of Clinical Immunology, Baltimore, MD, USA
| | - Minh Tran
- Johns Hopkins School of Medicine, Division of Clinical Immunology, Baltimore, MD, USA
| | | | | | - Bradley J Undem
- Johns Hopkins School of Medicine, Division of Clinical Immunology, Baltimore, MD, USA
| |
Collapse
|
9
|
Grisk O. Action potentials in postganglionic sympathetic nerves depend on Na V1.7. J Physiol 2024; 602:3239-3240. [PMID: 38805185 DOI: 10.1113/jp286827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 05/12/2024] [Indexed: 05/29/2024] Open
Affiliation(s)
- Olaf Grisk
- Institute of Physiology, Brandenburg Medical School Theodor Fontane, Neuruppin, Brandenburg, Germany
| |
Collapse
|
10
|
Xie YF, Yang J, Ratté S, Prescott SA. Similar excitability through different sodium channels and implications for the analgesic efficacy of selective drugs. eLife 2024; 12:RP90960. [PMID: 38687187 PMCID: PMC11060714 DOI: 10.7554/elife.90960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024] Open
Abstract
Nociceptive sensory neurons convey pain-related signals to the CNS using action potentials. Loss-of-function mutations in the voltage-gated sodium channel NaV1.7 cause insensitivity to pain (presumably by reducing nociceptor excitability) but clinical trials seeking to treat pain by inhibiting NaV1.7 pharmacologically have struggled. This may reflect the variable contribution of NaV1.7 to nociceptor excitability. Contrary to claims that NaV1.7 is necessary for nociceptors to initiate action potentials, we show that nociceptors can achieve similar excitability using different combinations of NaV1.3, NaV1.7, and NaV1.8. Selectively blocking one of those NaV subtypes reduces nociceptor excitability only if the other subtypes are weakly expressed. For example, excitability relies on NaV1.8 in acutely dissociated nociceptors but responsibility shifts to NaV1.7 and NaV1.3 by the fourth day in culture. A similar shift in NaV dependence occurs in vivo after inflammation, impacting ability of the NaV1.7-selective inhibitor PF-05089771 to reduce pain in behavioral tests. Flexible use of different NaV subtypes exemplifies degeneracy - achieving similar function using different components - and compromises reliable modulation of nociceptor excitability by subtype-selective inhibitors. Identifying the dominant NaV subtype to predict drug efficacy is not trivial. Degeneracy at the cellular level must be considered when choosing drug targets at the molecular level.
Collapse
Affiliation(s)
- Yu-Feng Xie
- Neurosciences and Mental Health, The Hospital for Sick ChildrenTorontoCanada
| | - Jane Yang
- Neurosciences and Mental Health, The Hospital for Sick ChildrenTorontoCanada
- Institute of Biomedical Engineering, University of TorontoTorontoCanada
| | - Stéphanie Ratté
- Neurosciences and Mental Health, The Hospital for Sick ChildrenTorontoCanada
| | - Steven A Prescott
- Neurosciences and Mental Health, The Hospital for Sick ChildrenTorontoCanada
- Institute of Biomedical Engineering, University of TorontoTorontoCanada
- Department of Physiology, University of TorontoTorontoCanada
| |
Collapse
|
11
|
Regan CP, Morissette P, Kraus RL, Wang E, Arrington L, Vavrek M, de Hoon J, Depre M, Lodeweyck T, Demeyer I, Laethem T, Stoch A, Struyk A. Autonomic Dysfunction Linked to Inhibition of the Na v1.7 Sodium Channel. Circulation 2024; 149:1394-1396. [PMID: 38648272 PMCID: PMC11027978 DOI: 10.1161/circulationaha.123.067331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Affiliation(s)
| | - Pierre Morissette
- Nonclinical Drug Safety (C.P.R., P.M., E.W.), Merck & Co., Inc., West Point, PA
| | | | - Erjia Wang
- Nonclinical Drug Safety (C.P.R., P.M., E.W.), Merck & Co., Inc., West Point, PA
| | - Leticia Arrington
- Pharmacokinetics, Pharmacodynamics and Drug Metabolism (L.A., M.V.), Merck & Co., Inc., West Point, PA
| | - Marissa Vavrek
- Pharmacokinetics, Pharmacodynamics and Drug Metabolism (L.A., M.V.), Merck & Co., Inc., West Point, PA
| | - Jan de Hoon
- Center for Clinical Pharmacology, Department of Pharmaceutical and Pharmacological Sciences, University of Leuven; Belgium (J.d.H., M.D., T. Lodeweyck)
| | - Marleen Depre
- Center for Clinical Pharmacology, Department of Pharmaceutical and Pharmacological Sciences, University of Leuven; Belgium (J.d.H., M.D., T. Lodeweyck)
| | - Thomas Lodeweyck
- Center for Clinical Pharmacology, Department of Pharmaceutical and Pharmacological Sciences, University of Leuven; Belgium (J.d.H., M.D., T. Lodeweyck)
| | - Ignace Demeyer
- Burn Center, Military Hospital Queen Astrid, Brussels, Belgium (I.D.)
| | - Tine Laethem
- Translational Medicine, Merck & Co., Inc., Upper Gwynedd, PA (T. Laethem, A. Stoch, A. Struyk)
| | - Aubrey Stoch
- Translational Medicine, Merck & Co., Inc., Upper Gwynedd, PA (T. Laethem, A. Stoch, A. Struyk)
| | - Arie Struyk
- Translational Medicine, Merck & Co., Inc., Upper Gwynedd, PA (T. Laethem, A. Stoch, A. Struyk)
| |
Collapse
|
12
|
Deng L, Dourado M, Reese RM, Huang K, Shields SD, Stark KL, Maksymetz J, Lin H, Kaminker JS, Jung M, Foreman O, Tao J, Ngu H, Joseph V, Roose-Girma M, Tam L, Lardell S, Orrhult LS, Karila P, Allard J, Hackos DH. Nav1.7 is essential for nociceptor action potentials in the mouse in a manner independent of endogenous opioids. Neuron 2023; 111:2642-2659.e13. [PMID: 37352856 DOI: 10.1016/j.neuron.2023.05.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 04/07/2023] [Accepted: 05/26/2023] [Indexed: 06/25/2023]
Abstract
Loss-of-function mutations in Nav1.7, a voltage-gated sodium channel, cause congenital insensitivity to pain (CIP) in humans, demonstrating that Nav1.7 is essential for the perception of pain. However, the mechanism by which loss of Nav1.7 results in insensitivity to pain is not entirely clear. It has been suggested that loss of Nav1.7 induces overexpression of enkephalin, an endogenous opioid receptor agonist, leading to opioid-dependent analgesia. Using behavioral pharmacology and single-cell RNA-seq analysis, we find that overexpression of enkephalin occurs only in cLTMR neurons, a subclass of sensory neurons involved in low-threshold touch detection, and that this overexpression does not play a role in the analgesia observed following genetic removal of Nav1.7. Furthermore, we demonstrate using laser speckle contrast imaging (LSCI) and in vivo electrophysiology that Nav1.7 function is required for the initiation of C-fiber action potentials (APs), which explains the observed insensitivity to pain following genetic removal or inhibition of Nav1.7.
Collapse
Affiliation(s)
- Lunbin Deng
- Department of Neuroscience, Genentech, Inc., 1 DNA Way, South San Francisco, CA, USA
| | - Michelle Dourado
- Department of Neuroscience, Genentech, Inc., 1 DNA Way, South San Francisco, CA, USA
| | - Rebecca M Reese
- Department of Neuroscience, Genentech, Inc., 1 DNA Way, South San Francisco, CA, USA
| | - Kevin Huang
- Department of OMNI Bioinformatics, Genentech, Inc., 1 DNA Way, South San Francisco, CA, USA
| | - Shannon D Shields
- Department of Neuroscience, Genentech, Inc., 1 DNA Way, South San Francisco, CA, USA
| | - Kimberly L Stark
- Department of Neuroscience, Genentech, Inc., 1 DNA Way, South San Francisco, CA, USA
| | - James Maksymetz
- Department of Neuroscience, Genentech, Inc., 1 DNA Way, South San Francisco, CA, USA
| | - Han Lin
- Department of Neuroscience, Genentech, Inc., 1 DNA Way, South San Francisco, CA, USA
| | - Joshua S Kaminker
- Department of OMNI Bioinformatics, Genentech, Inc., 1 DNA Way, South San Francisco, CA, USA
| | - Min Jung
- Department of OMNI Bioinformatics, Genentech, Inc., 1 DNA Way, South San Francisco, CA, USA
| | - Oded Foreman
- Department of Pathology, Genentech, Inc., 1 DNA Way, South San Francisco, CA, USA
| | - Janet Tao
- Department of Pathology, Genentech, Inc., 1 DNA Way, South San Francisco, CA, USA
| | - Hai Ngu
- Department of Pathology, Genentech, Inc., 1 DNA Way, South San Francisco, CA, USA
| | - Victory Joseph
- Department of Biomedical Imaging, Genentech, Inc., 1 DNA Way, South San Francisco, CA, USA
| | - Meron Roose-Girma
- Department of Molecular Biology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Lucinda Tam
- Department of Molecular Biology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | | | | | - Paul Karila
- Cellectricon AB, Neongatan 4B, 431 53 Mölndal, Sweden
| | - Julien Allard
- E-Phys, CRBC, 28 place Henri Dunant, 63000 Clermont-Ferrand, France.
| | - David H Hackos
- Department of Neuroscience, Genentech, Inc., 1 DNA Way, South San Francisco, CA, USA.
| |
Collapse
|
13
|
Kitano Y, Shinozuka T. Inhibition of Na V1.7: the possibility of ideal analgesics. RSC Med Chem 2022; 13:895-920. [PMID: 36092147 PMCID: PMC9384491 DOI: 10.1039/d2md00081d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 07/25/2022] [Indexed: 08/03/2023] Open
Abstract
The selective inhibition of NaV1.7 is a promising strategy for developing novel analgesic agents with fewer adverse effects. Although the potent selective inhibition of NaV1.7 has been recently achieved, multiple NaV1.7 inhibitors failed in clinical development. In this review, the relationship between preclinical in vivo efficacy and NaV1.7 coverage among three types of voltage-gated sodium channel (VGSC) inhibitors, namely conventional VGSC inhibitors, sulphonamides and acyl sulphonamides, is discussed. By demonstrating the PK/PD discrepancy of preclinical studies versus in vivo models and clinical results, the potential reasons behind the disconnect between preclinical results and clinical outcomes are discussed together with strategies for developing ideal analgesic agents.
Collapse
Affiliation(s)
- Yutaka Kitano
- R&D Division, Daiichi Sankyo Co., Ltd. 1-2-58 Hiromachi Shinagawa-ku Tokyo 140-8710 Japan
| | - Tsuyoshi Shinozuka
- R&D Division, Daiichi Sankyo Co., Ltd. 1-2-58 Hiromachi Shinagawa-ku Tokyo 140-8710 Japan
| |
Collapse
|
14
|
Jiang D, Zhang J, Xia Z. Structural Advances in Voltage-Gated Sodium Channels. Front Pharmacol 2022; 13:908867. [PMID: 35721169 PMCID: PMC9204039 DOI: 10.3389/fphar.2022.908867] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/23/2022] [Indexed: 11/17/2022] Open
Abstract
Voltage-gated sodium (NaV) channels are responsible for the rapid rising-phase of action potentials in excitable cells. Over 1,000 mutations in NaV channels are associated with human diseases including epilepsy, periodic paralysis, arrhythmias and pain disorders. Natural toxins and clinically-used small-molecule drugs bind to NaV channels and modulate their functions. Recent advances from cryo-electron microscopy (cryo-EM) structures of NaV channels reveal invaluable insights into the architecture, activation, fast inactivation, electromechanical coupling, ligand modulation and pharmacology of eukaryotic NaV channels. These structural analyses not only demonstrate molecular mechanisms for NaV channel structure and function, but also provide atomic level templates for rational development of potential subtype-selective therapeutics. In this review, we summarize recent structural advances of eukaryotic NaV channels, highlighting the structural features of eukaryotic NaV channels as well as distinct modulation mechanisms by a wide range of modulators from natural toxins to synthetic small-molecules.
Collapse
Affiliation(s)
- Daohua Jiang
- Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- *Correspondence: Daohua Jiang,
| | - Jiangtao Zhang
- Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Molecular Biophysics of MOE, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Zhanyi Xia
- Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
15
|
Wisedchaisri G, Gamal El-Din TM. Druggability of Voltage-Gated Sodium Channels-Exploring Old and New Drug Receptor Sites. Front Pharmacol 2022; 13:858348. [PMID: 35370700 PMCID: PMC8968173 DOI: 10.3389/fphar.2022.858348] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 03/01/2022] [Indexed: 01/12/2023] Open
Abstract
Voltage-gated ion channels are important drug targets because they play crucial physiological roles in both excitable and non-excitable cells. About 15% of clinical drugs used for treating human diseases target ion channels. However, most of these drugs do not provide sufficient specificity to a single subtype of the channels and their off-target side effects can be serious and sometimes fatal. Recent advancements in imaging techniques have enabled us for the first time to visualize unique and hidden parts of voltage-gated sodium channels in different structural conformations, and to develop drugs that further target a selected functional state in each channel subtype with the potential for high precision and low toxicity. In this review we describe the druggability of voltage-gated sodium channels in distinct functional states, which could potentially be used to selectively target the channels. We review classical drug receptors in the channels that have recently been structurally characterized by cryo-electron microscopy with natural neurotoxins and clinical drugs. We further examine recent drug discoveries for voltage-gated sodium channels and discuss opportunities to use distinct, state-dependent receptor sites in the voltage sensors as unique drug targets. Finally, we explore potential new receptor sites that are currently unknown for sodium channels but may be valuable for future drug discovery. The advancement presented here will help pave the way for drug development that selectively targets voltage-gated sodium channels.
Collapse
Affiliation(s)
- Goragot Wisedchaisri
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Tamer M Gamal El-Din
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| |
Collapse
|
16
|
Brown DG, Wobst HJ. A survey of the clinical pipeline in neuroscience. Bioorg Med Chem Lett 2022; 56:128482. [PMID: 34864194 DOI: 10.1016/j.bmcl.2021.128482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 11/21/2021] [Accepted: 11/22/2021] [Indexed: 11/02/2022]
Abstract
Many new first-in-class drugs for neuroscience indications have been introduced in the past decade including new treatments for migraine, amyotrophic lateral sclerosis, depression, and multiple sclerosis. However, significant unmet patient needs remain in areas such as chronic pain, neurodegeneration, psychiatric diseases, and epilepsy. This review summarizes some of the advanced clinical compounds for these indications. Additionally, current opportunities and challenges that remain with respect to genetic validation, biomarkers, and translational models are discussed.
Collapse
Affiliation(s)
- Dean G Brown
- Jnana Therapeutics, 6 Tide St, MA 02210, United States.
| | - Heike J Wobst
- Jnana Therapeutics, 6 Tide St, MA 02210, United States
| |
Collapse
|
17
|
Ballard JE, Pall PS, Vardigan J, Zhao F, Holahan MA, Zhou X, Jochnowitz N, Kraus RL, Klein RM, Henze DA, Houghton AK, Burgey CS, Gibson C, Struyk A. Translational Pharmacokinetic–Pharmacodynamic Modeling of NaV1.7 Inhibitor MK-2075 to Inform Human Efficacious Dose. Front Pharmacol 2021; 12:786078. [PMID: 35002718 PMCID: PMC8740778 DOI: 10.3389/fphar.2021.786078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 11/17/2021] [Indexed: 11/21/2022] Open
Abstract
MK-2075 is a small-molecule selective inhibitor of the NaV1.7 channel investigated for the treatment of postoperative pain. A translational strategy was developed for MK-2075 to quantitatively interrelate drug exposure, target modulation, and the desired pharmacological response in preclinical animal models for the purpose of human translation. Analgesics used as a standard of care in postoperative pain were evaluated in preclinical animal models of nociceptive behavior (mouse tail flick latency and rhesus thermode heat withdrawal) to determine the magnitude of pharmacodynamic (PD) response at plasma concentrations associated with efficacy in the clinic. MK-2075 was evaluated in those same animal models to determine the concentration of MK-2075 required to achieve the desired level of response. Translation of MK-2075 efficacious concentrations in preclinical animal models to a clinical PKPD target in humans was achieved by accounting for species differences in plasma protein binding and in vitro potency against the NaV1.7 channel. Estimates of human pharmacokinetic (PK) parameters were obtained from allometric scaling of a PK model from preclinical species and used to predict the dose required to achieve the clinical exposure. MK-2075 exposure–response in a preclinical target modulation assay (rhesus olfaction) was characterized using a computational PKPD model which included a biophase compartment to account for the observed hysteresis. Translation of this model to humans was accomplished by correcting for species differences in PK NaV1.7 potency, and plasma protein binding while assuming that the kinetics of distribution to the target site is the same between humans and rhesus monkeys. This enabled prediction of the level of target modulation anticipated to be achieved over the dosing interval at the projected clinical efficacious human dose. Integration of these efforts into the early development plan informed clinical study design and decision criteria.
Collapse
Affiliation(s)
- Jeanine E. Ballard
- Pharmacokinetics Pharmacodynamics and Drug Metabolism, Merck & Co. Inc., Kenilworth, NJ, United States
- *Correspondence: Jeanine E. Ballard,
| | - Parul S. Pall
- Neuroscience Pharmacology, Merck & Co. Inc., Kenilworth, NJ, United States
| | - Joshua Vardigan
- Neuroscience Pharmacology, Merck & Co. Inc., Kenilworth, NJ, United States
| | - Fuqiang Zhao
- Translational Imaging Biomarkers, Merck & Co. Inc., Kenilworth, NJ, United States
| | - Marie A. Holahan
- Translational Imaging Biomarkers, Merck & Co. Inc., Kenilworth, NJ, United States
| | - Xiaoping Zhou
- Neuroscience Pharmacology, Merck & Co. Inc., Kenilworth, NJ, United States
| | - Nina Jochnowitz
- Neuroscience Pharmacology, Merck & Co. Inc., Kenilworth, NJ, United States
| | - Richard L. Kraus
- Neuroscience Pharmacology, Merck & Co. Inc., Kenilworth, NJ, United States
| | - Rebecca M. Klein
- Neuroscience Pharmacology, Merck & Co. Inc., Kenilworth, NJ, United States
| | - Darrell A. Henze
- Neuroscience Pharmacology, Merck & Co. Inc., Kenilworth, NJ, United States
| | - Andrea K. Houghton
- Neuroscience Pharmacology, Merck & Co. Inc., Kenilworth, NJ, United States
| | | | - Christopher Gibson
- Pharmacokinetics Pharmacodynamics and Drug Metabolism, Merck & Co. Inc., Kenilworth, NJ, United States
| | - Arie Struyk
- Translational Medicine, Merck & Co. Inc., Kenilworth, NJ, United States
| |
Collapse
|
18
|
Cai S, Moutal A, Yu J, Chew LA, Isensee J, Chawla R, Gomez K, Luo S, Zhou Y, Chefdeville A, Madura C, Perez-Miller S, Bellampalli SS, Dorame A, Scott DD, François-Moutal L, Shan Z, Woodward T, Gokhale V, Hohmann AG, Vanderah TW, Patek M, Khanna M, Hucho T, Khanna R. Selective targeting of NaV1.7 via inhibition of the CRMP2-Ubc9 interaction reduces pain in rodents. Sci Transl Med 2021; 13:eabh1314. [PMID: 34757807 PMCID: PMC11729770 DOI: 10.1126/scitranslmed.abh1314] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The voltage-gated sodium NaV1.7 channel, critical for sensing pain, has been actively targeted by drug developers; however, there are currently no effective and safe therapies targeting NaV1.7. Here, we tested whether a different approach, indirect NaV1.7 regulation, could have antinociceptive effects in preclinical models. We found that preventing addition of small ubiquitin-like modifier (SUMO) on the NaV1.7-interacting cytosolic collapsin response mediator protein 2 (CRMP2) blocked NaV1.7 functions and had antinociceptive effects in rodents. In silico targeting of the SUMOylation site in CRMP2 (Lys374) identified >200 hits, of which compound 194 exhibited selective in vitro and ex vivo NaV1.7 engagement. Orally administered 194 was not only antinociceptive in preclinical models of acute and chronic pain but also demonstrated synergy alongside other analgesics—without eliciting addiction, rewarding properties, or neurotoxicity. Analgesia conferred by 194 was opioid receptor dependent. Our results demonstrate that 194 is a first-in-class protein-protein inhibitor that capitalizes on CRMP2-NaV1.7 regulation to deliver safe analgesia in rodents.
Collapse
Affiliation(s)
- Song Cai
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Aubin Moutal
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Jie Yu
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Lindsey A. Chew
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Jörg Isensee
- Department of Anesthesiology and Intensive Care Medicine, Translational Pain Research, University Hospital of Cologne, University Cologne, Joseph-Stelzmann-Str 9, Cologne D-50931, Germany
| | - Reena Chawla
- BIO5 Institute, 1657 East Helen Street, Tucson, AZ 85721, USA
| | - Kimberly Gomez
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Shizhen Luo
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Yuan Zhou
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Aude Chefdeville
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Cynthia Madura
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Samantha Perez-Miller
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
- Center for Innovation in Brain Sciences, University of Arizona, Tucson, AZ 85721, USA
| | - Shreya Sai Bellampalli
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Angie Dorame
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - David D. Scott
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Liberty François-Moutal
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Zhiming Shan
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Taylor Woodward
- Department of Psychological and Brain Sciences, Program in Neuroscience and Gill Center for Biomolecular Science, Indiana University, Bloomington, IN 47405-2204, USA
| | - Vijay Gokhale
- BIO5 Institute, 1657 East Helen Street, Tucson, AZ 85721, USA
- College of Pharmacy, University of Arizona, 1703 East Mabel Street, Tucson, AZ 85721, USA
| | - Andrea G. Hohmann
- Department of Psychological and Brain Sciences, Program in Neuroscience and Gill Center for Biomolecular Science, Indiana University, Bloomington, IN 47405-2204, USA
| | - Todd W. Vanderah
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
- Comprehensive Pain and Addiction Center, The University of Arizona, Tucson, AZ 85724, USA
| | - Marcel Patek
- Regulonix LLC, 1555 E. Entrada Segunda, Tucson, AZ 85718, USA
- Bright Rock Path LLC, Tucson, AZ 85724, USA
| | - May Khanna
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
- BIO5 Institute, 1657 East Helen Street, Tucson, AZ 85721, USA
- Center for Innovation in Brain Sciences, University of Arizona, Tucson, AZ 85721, USA
- Regulonix LLC, 1555 E. Entrada Segunda, Tucson, AZ 85718, USA
| | - Tim Hucho
- Department of Anesthesiology and Intensive Care Medicine, Translational Pain Research, University Hospital of Cologne, University Cologne, Joseph-Stelzmann-Str 9, Cologne D-50931, Germany
| | - Rajesh Khanna
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
- BIO5 Institute, 1657 East Helen Street, Tucson, AZ 85721, USA
- Center for Innovation in Brain Sciences, University of Arizona, Tucson, AZ 85721, USA
- Comprehensive Pain and Addiction Center, The University of Arizona, Tucson, AZ 85724, USA
- Regulonix LLC, 1555 E. Entrada Segunda, Tucson, AZ 85718, USA
| |
Collapse
|
19
|
Yu L, Tsuji K, Ujihara I, Liu Q, Pavelkova N, Tsujimura T, Inoue M, Meeker S, Nisenbaum E, McDermott JS, Krajewski J, Undem BJ, Kollarik M, Canning BJ. Antitussive effects of Na V 1.7 blockade in Guinea pigs. Eur J Pharmacol 2021; 907:174192. [PMID: 34010618 DOI: 10.1016/j.ejphar.2021.174192] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 05/07/2021] [Accepted: 05/12/2021] [Indexed: 01/25/2023]
Abstract
Our previous studies implicated the voltage-gated sodium channel subtype NaV 1.7 in the transmission of action potentials by the vagal afferent nerves regulating cough and thus identified this channel as a rational therapeutic target for antitussive therapy. But it is presently unclear whether a systemically administered small molecule inhibitor of NaV 1.7 conductance can achieve therapeutic benefit in the absence of side effects on cardiovascular function, gastrointestinal motility or respiration. To this end, we have evaluated the antitussive effects of the NaV 1.7 selective blocker Compound 801 administered systemically in awake guinea pigs or administered topically in anesthetized guinea pigs. We also evaluated the antitussive effects of ambroxol, a low affinity NaV blocker modestly selective for tetrodotoxin resistant NaV subtypes. Both Compound 801 and ambroxol dose-dependently inhibited action potential conduction in guinea pig vagus nerves (assessed by compound potential), with ambroxol nearly 100-fold less potent than the NaV 1.7 selective Compound 801 in this and other NaV 1.7-dependent guinea pig and human tissue-based assays. Both drugs also inhibited citric acid evoked coughing in awake or anesthetized guinea pigs, with potencies supportive of an NaV 1.7-dependent mechanism. Notably, however, the antitussive effects of systemically administered Compound 801 were accompanied by hypotension and respiratory depression. Given the antitussive effects of topically administered Compound 801, we speculate that the likely insurmountable side effects on blood pressure and respiratory drive associated with systemic dosing make topical formulations a viable and perhaps unavoidable therapeutic strategy for targeting NaV 1.7 in cough.
Collapse
Affiliation(s)
- Li Yu
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongii University School of Medicine, Shanghai, 200065, China
| | - Kojun Tsuji
- Division of Dysphagia Rehabilitation, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Izumi Ujihara
- Division of Dysphagia Rehabilitation, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Qi Liu
- Johns Hopkins Asthma and Allergy Center, 5501 Hopkins Bayview Circle, Baltimore, MD, 21224, USA
| | - Nikoleta Pavelkova
- Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Takanori Tsujimura
- Division of Dysphagia Rehabilitation, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Makoto Inoue
- Division of Dysphagia Rehabilitation, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Sonya Meeker
- Johns Hopkins Asthma and Allergy Center, 5501 Hopkins Bayview Circle, Baltimore, MD, 21224, USA
| | - Eric Nisenbaum
- Lilly Research Laboratories, Indianapolis, IN, 46285, USA
| | | | - Jeff Krajewski
- Lilly Research Laboratories, Indianapolis, IN, 46285, USA
| | - Bradley J Undem
- Johns Hopkins Asthma and Allergy Center, 5501 Hopkins Bayview Circle, Baltimore, MD, 21224, USA
| | - Marian Kollarik
- Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Brendan J Canning
- Johns Hopkins Asthma and Allergy Center, 5501 Hopkins Bayview Circle, Baltimore, MD, 21224, USA.
| |
Collapse
|
20
|
Roecker AJ, Layton ME, Pero JE, Kelly MJ, Greshock TJ, Kraus RL, Li Y, Klein R, Clements M, Daley C, Jovanovska A, Ballard JE, Wang D, Zhao F, Brunskill APJ, Peng X, Wang X, Sun H, Houghton AK, Burgey CS. Discovery of Arylsulfonamide Na v1.7 Inhibitors: IVIVC, MPO Methods, and Optimization of Selectivity Profile. ACS Med Chem Lett 2021; 12:1038-1049. [PMID: 34141090 PMCID: PMC8201757 DOI: 10.1021/acsmedchemlett.1c00218] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 05/26/2021] [Indexed: 01/13/2023] Open
Abstract
The voltage-gated sodium channel Nav1.7 continues to be a high-profile target for the treatment of various pain afflictions due to its strong human genetic validation. While isoform selective molecules have been discovered and advanced into the clinic, to date, this target has yet to bear fruit in the form of marketed therapeutics for the treatment of pain. Lead optimization efforts over the past decade have focused on selectivity over Nav1.5 due to its link to cardiac side effects as well as the translation of preclinical efficacy to man. Inhibition of Nav1.6 was recently reported to yield potential respiratory side effects preclinically, and this finding necessitated a modified target selectivity profile. Herein, we report the continued optimization of a novel series of arylsulfonamide Nav1.7 inhibitors to afford improved selectivity over Nav1.6 while maintaining rodent oral bioavailability through the use of a novel multiparameter optimization (MPO) paradigm. We also report in vitro-in vivo correlations from Nav1.7 electrophysiology protocols to preclinical models of efficacy to assist in projecting clinical doses. These efforts produced inhibitors such as compound 19 with potency against Nav1.7, selectivity over Nav1.5 and Nav1.6, and efficacy in behavioral models of pain in rodents as well as inhibition of rhesus olfactory response indicative of target modulation.
Collapse
Affiliation(s)
- Anthony J. Roecker
- Discovery
Chemistry, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Mark E. Layton
- Discovery
Chemistry, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Joseph E. Pero
- Discovery
Chemistry, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Michael J. Kelly
- Discovery
Chemistry, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Thomas J. Greshock
- Discovery
Chemistry, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Richard L. Kraus
- Pharmacology, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Yuxing Li
- Pharmacology, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Rebecca Klein
- Pharmacology, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Michelle Clements
- Pharmacology, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Christopher Daley
- Pharmacology, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Aneta Jovanovska
- Pharmacology, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Jeanine E. Ballard
- Pharmacokinetic,
Pharmacodynamics, and Drug Metabolism, Merck
& Co., Inc., West Point, Pennsylvania 19486, United States
| | - Deping Wang
- Computational
and Structural Chemistry, Merck & Co.,
Inc., West Point, Pennsylvania 19486, United States
| | - Fuqiang Zhao
- Translational
Imaging and Biomarkers, Merck & Co.,
Inc., West Point, Pennsylvania 19486, United States
| | - Andrew P. J. Brunskill
- Molecular
and Materials Characterization, Merck &
Co., Inc., Rahway, New Jersey 07065, United States
| | - Xuanjia Peng
- HitS
Unite, WuXi AppTec Co., Ltd. (Shanghai), Shanghai 200131, China
| | - Xiu Wang
- IDSU, WuXi AppTec
Co., Ltd. (Shanghai), Shanghai 200131, China
| | - Haiyan Sun
- IDSU, WuXi AppTec
Co., Ltd. (Shanghai), Shanghai 200131, China
| | - Andrea K. Houghton
- Pharmacology, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Christopher S. Burgey
- Discovery
Chemistry, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| |
Collapse
|
21
|
Safina BS, McKerrall SJ, Sun S, Chen CA, Chowdhury S, Jia Q, Li J, Zenova AY, Andrez JC, Bankar G, Bergeron P, Chang JH, Chang E, Chen J, Dean R, Decker SM, DiPasquale A, Focken T, Hemeon I, Khakh K, Kim A, Kwan R, Lindgren A, Lin S, Maher J, Mezeyova J, Misner D, Nelkenbrecher K, Pang J, Reese R, Shields SD, Sojo L, Sheng T, Verschoof H, Waldbrook M, Wilson MS, Xie Z, Young C, Zabka TS, Hackos DH, Ortwine DF, White AD, Johnson JP, Robinette CL, Dehnhardt CM, Cohen CJ, Sutherlin DP. Discovery of Acyl-sulfonamide Na v1.7 Inhibitors GDC-0276 and GDC-0310. J Med Chem 2021; 64:2953-2966. [PMID: 33682420 DOI: 10.1021/acs.jmedchem.1c00049] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nav1.7 is an extensively investigated target for pain with a strong genetic link in humans, yet in spite of this effort, it remains challenging to identify efficacious, selective, and safe inhibitors. Here, we disclose the discovery and preclinical profile of GDC-0276 (1) and GDC-0310 (2), selective Nav1.7 inhibitors that have completed Phase 1 trials. Our initial search focused on close-in analogues to early compound 3. This resulted in the discovery of GDC-0276 (1), which possessed improved metabolic stability and an acceptable overall pharmacokinetics profile. To further derisk the predicted human pharmacokinetics and enable QD dosing, additional optimization of the scaffold was conducted, resulting in the discovery of a novel series of N-benzyl piperidine Nav1.7 inhibitors. Improvement of the metabolic stability by blocking the labile benzylic position led to the discovery of GDC-0310 (2), which possesses improved Nav selectivity and pharmacokinetic profile over 1.
Collapse
Affiliation(s)
- Brian S Safina
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Steven J McKerrall
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Shaoyi Sun
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Chien-An Chen
- Chempartner, Building No. 5, 998 Halei Road, Zhangjiang Hi-Tech Park, Pudong New Area, Shanghai 201203, P.R. China
| | - Sultan Chowdhury
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Qi Jia
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Jun Li
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Alla Y Zenova
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Jean-Christophe Andrez
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Girish Bankar
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Philippe Bergeron
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Jae H Chang
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Elaine Chang
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Jun Chen
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Richard Dean
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Shannon M Decker
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Antonio DiPasquale
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Thilo Focken
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Ivan Hemeon
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Kuldip Khakh
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Amy Kim
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Rainbow Kwan
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Andrea Lindgren
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Sophia Lin
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Jonathan Maher
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Janette Mezeyova
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Dinah Misner
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Karen Nelkenbrecher
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Jodie Pang
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Rebecca Reese
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Shannon D Shields
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Luis Sojo
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Tao Sheng
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Henry Verschoof
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Matthew Waldbrook
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Michael S Wilson
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Zhiwei Xie
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Clint Young
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Tanja S Zabka
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - David H Hackos
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Daniel F Ortwine
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Andrew D White
- Chempartner, Building No. 5, 998 Halei Road, Zhangjiang Hi-Tech Park, Pudong New Area, Shanghai 201203, P.R. China
| | - J P Johnson
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - C Lee Robinette
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Christoph M Dehnhardt
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Charles J Cohen
- Xenon Pharmaceuticals, Inc., 200-3650 Gilmore Way, Burnaby, British Columbia V5G 4W8, Canada
| | - Daniel P Sutherlin
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
22
|
Eagles DA, Chow CY, King GF. Fifteen years of Na
V
1.7 channels as an analgesic target: Why has excellent in vitro pharmacology not translated into in vivo analgesic efficacy? Br J Pharmacol 2020; 179:3592-3611. [DOI: 10.1111/bph.15327] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/14/2020] [Accepted: 10/23/2020] [Indexed: 12/16/2022] Open
Affiliation(s)
- David A. Eagles
- Institute for Molecular Bioscience The University of Queensland St Lucia QLD Australia
| | - Chun Yuen Chow
- Institute for Molecular Bioscience The University of Queensland St Lucia QLD Australia
| | - Glenn F. King
- Institute for Molecular Bioscience The University of Queensland St Lucia QLD Australia
| |
Collapse
|
23
|
Alsaloum M, Higerd GP, Effraim PR, Waxman SG. Status of peripheral sodium channel blockers for non-addictive pain treatment. Nat Rev Neurol 2020; 16:689-705. [PMID: 33110213 DOI: 10.1038/s41582-020-00415-2] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2020] [Indexed: 12/15/2022]
Abstract
The effective and safe treatment of pain is an unmet health-care need. Current medications used for pain management are often only partially effective, carry dose-limiting adverse effects and are potentially addictive, highlighting the need for improved therapeutic agents. Most common pain conditions originate in the periphery, where dorsal root ganglion and trigeminal ganglion neurons feed pain information into the CNS. Voltage-gated sodium (NaV) channels drive neuronal excitability and three subtypes - NaV1.7, NaV1.8 and NaV1.9 - are preferentially expressed in the peripheral nervous system, suggesting that their inhibition might treat pain while avoiding central and cardiac adverse effects. Genetic and functional studies of human pain disorders have identified NaV1.7, NaV1.8 and NaV1.9 as mediators of pain and validated them as targets for pain treatment. Consequently, multiple NaV1.7-specific and NaV1.8-specific blockers have undergone clinical trials, with others in preclinical development, and the targeting of NaV1.9, although hampered by technical constraints, might also be moving ahead. In this Review, we summarize the clinical and preclinical literature describing compounds that target peripheral NaV channels and discuss the challenges and future prospects for the field. Although the potential of peripheral NaV channel inhibition for the treatment of pain has yet to be realized, this remains a promising strategy to achieve non-addictive analgesia for multiple pain conditions.
Collapse
Affiliation(s)
- Matthew Alsaloum
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.,Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, USA.,Yale Medical Scientist Training Program, Yale School of Medicine, New Haven, CT, USA.,Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT, USA
| | - Grant P Higerd
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.,Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, USA.,Yale Medical Scientist Training Program, Yale School of Medicine, New Haven, CT, USA
| | - Philip R Effraim
- Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, USA.,Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA. .,Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA. .,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, USA.
| |
Collapse
|
24
|
Pajouhesh H, Beckley JT, Delwig A, Hajare HS, Luu G, Monteleone D, Zhou X, Ligutti J, Amagasu S, Moyer BD, Yeomans DC, Du Bois J, Mulcahy JV. Discovery of a selective, state-independent inhibitor of Na V1.7 by modification of guanidinium toxins. Sci Rep 2020; 10:14791. [PMID: 32908170 PMCID: PMC7481244 DOI: 10.1038/s41598-020-71135-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 08/06/2020] [Indexed: 12/15/2022] Open
Abstract
The voltage-gated sodium channel isoform NaV1.7 is highly expressed in dorsal root ganglion neurons and is obligatory for nociceptive signal transmission. Genetic gain-of-function and loss-of-function NaV1.7 mutations have been identified in select individuals, and are associated with episodic extreme pain disorders and insensitivity to pain, respectively. These findings implicate NaV1.7 as a key pharmacotherapeutic target for the treatment of pain. While several small molecules targeting NaV1.7 have been advanced to clinical development, no NaV1.7-selective compound has shown convincing efficacy in clinical pain applications. Here we describe the discovery and characterization of ST-2262, a NaV1.7 inhibitor that blocks the extracellular vestibule of the channel with an IC50 of 72 nM and greater than 200-fold selectivity over off-target sodium channel isoforms, NaV1.1-1.6 and NaV1.8. In contrast to other NaV1.7 inhibitors that preferentially inhibit the inactivated state of the channel, ST-2262 is equipotent in a protocol that favors the resting state of the channel, a protocol that favors the inactivated state, and a high frequency protocol. In a non-human primate study, animals treated with ST-2262 exhibited reduced sensitivity to noxious heat. These findings establish the extracellular vestibule of the sodium channel as a viable receptor site for the design of selective ligands targeting NaV1.7.
Collapse
Affiliation(s)
- H Pajouhesh
- SiteOne Therapeutics, South San Francisco, CA, 94080, USA
| | - J T Beckley
- SiteOne Therapeutics, Bozeman, MT, 59715, USA
| | - A Delwig
- SiteOne Therapeutics, South San Francisco, CA, 94080, USA
| | - H S Hajare
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA
| | - G Luu
- SiteOne Therapeutics, South San Francisco, CA, 94080, USA
| | - D Monteleone
- SiteOne Therapeutics, South San Francisco, CA, 94080, USA
| | - X Zhou
- SiteOne Therapeutics, South San Francisco, CA, 94080, USA
| | - J Ligutti
- Neuroscience Department, Amgen Research, Thousand Oaks, CA, 91320, USA
| | - S Amagasu
- Neuroscience Department, Amgen Research, Thousand Oaks, CA, 91320, USA
| | - B D Moyer
- Neuroscience Department, Amgen Research, Thousand Oaks, CA, 91320, USA
| | - D C Yeomans
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University, Stanford, CA, 94305, USA
| | - J Du Bois
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA
| | - J V Mulcahy
- SiteOne Therapeutics, South San Francisco, CA, 94080, USA.
| |
Collapse
|
25
|
Application of Pharmacokinetic-Pharmacodynamic Modeling to Inform Translation of In Vitro NaV1.7 Inhibition to In Vivo Pharmacological Response in Non-human Primate. Pharm Res 2020; 37:181. [PMID: 32888082 PMCID: PMC7473964 DOI: 10.1007/s11095-020-02914-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 08/18/2020] [Indexed: 12/25/2022]
Abstract
Purpose This work describes a staged approach to the application of pharmacokinetic-pharmacodynamic (PK-PD) modeling in the voltage-gated sodium ion channel (NaV1.7) inhibitor drug discovery effort to address strategic questions regarding in vitro to in vivo translation of target modulation. Methods PK-PD analysis was applied to data from a functional magnetic resonance imaging (fMRI) technique to non-invasively measure treatment mediated inhibition of olfaction signaling in non-human primates (NHPs). Initial exposure-response was evaluated using single time point data pooled across 27 compounds to inform on in vitro to in vivo correlation (IVIVC). More robust effect compartment PK-PD modeling was conducted for a subset of 10 compounds with additional PD and PK data to characterize hysteresis. Results The pooled compound exposure-response facilitated an early exploration of IVIVC with a limited dataset for each individual compound, and it suggested a 2.4-fold in vitro to in vivo scaling factor for the NaV1.7 target. Accounting for hysteresis with an effect compartment PK-PD model as compounds advanced towards preclinical development provided a more robust determination of in vivo potency values, which resulted in a statistically significant positive IVIVC with a slope of 1.057 ± 0.210, R-squared of 0.7831, and p value of 0.006. Subsequent simulations with the PK-PD model informed the design of anti-nociception efficacy studies in NHPs. Conclusions A staged approach to PK-PD modeling and simulation enabled integration of in vitro NaV1.7 potency, plasma protein binding, and pharmacokinetics to describe the exposure-response profile and inform future study design as the NaV1.7 inhibitor effort progressed through drug discovery. Electronic supplementary material The online version of this article (10.1007/s11095-020-02914-9) contains supplementary material, which is available to authorized users.
Collapse
|
26
|
Grubinska B, Chen L, Alsaloum M, Rampal N, Matson DJ, Yang C, Taborn K, Zhang M, Youngblood B, Liu D, Galbreath E, Allred S, Lepherd M, Ferrando R, Kornecook TJ, Lehto SG, Waxman SG, Moyer BD, Dib-Hajj S, Gingras J. Rat Na V1.7 loss-of-function genetic model: Deficient nociceptive and neuropathic pain behavior with retained olfactory function and intra-epidermal nerve fibers. Mol Pain 2020; 15:1744806919881846. [PMID: 31550995 PMCID: PMC6831982 DOI: 10.1177/1744806919881846] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Recapitulating human disease pathophysiology using genetic animal models is a
powerful approach to enable mechanistic understanding of genotype–phenotype
relationships for drug development. NaV1.7 is a sodium channel
expressed in the peripheral nervous system with strong human genetic validation
as a pain target. Efforts to identify novel analgesics that are nonaddictive
resulted in industry exploration of a class of sulfonamide compounds that bind
to the fourth voltage-sensor domain of NaV1.7. Due to sequence
differences in this region, sulfonamide blockers generally are potent on human
but not rat NaV1.7 channels. To test sulfonamide-based chemical
matter in rat models of pain, we generated a humanized NaV1.7 rat
expressing a chimeric NaV1.7 protein containing the
sulfonamide-binding site of the human gene sequence as a replacement for the
equivalent rat sequence. Unexpectedly, upon transcription, the human insert was
spliced out, resulting in a premature stop codon. Using a validated antibody,
NaV1.7 protein was confirmed to be lost in the brainstem, dorsal
root ganglia, sciatic nerve, and gastrointestinal tissue but not in nasal
turbinates or olfactory bulb in rats homozygous for the knock-in allele
(HOM-KI). HOM-KI rats exhibited normal intraepidermal nerve fiber density with
reduced tetrodotoxin-sensitive current density and action potential firing in
small diameter dorsal root ganglia neurons. HOM-KI rats did not exhibit
nociceptive pain responses in hot plate or capsaicin-induced flinching assays
and did not exhibit neuropathic pain responses following spinal nerve ligation.
Consistent with expression of chimeric NaV1.7 in olfactory tissue,
HOM-KI rats retained olfactory function. This new genetic model highlights the
necessity of NaV1.7 for pain behavior in rats and indicates that
sufficient inhibition of NaV1.7 in humans may reduce pain in
neuropathic conditions. Due to preserved olfactory function, this rat model
represents an alternative to global NaV1.7 knockout mice that require
time-intensive hand feeding during early postnatal development.
Collapse
Affiliation(s)
- B Grubinska
- Neuroscience Department, Amgen Research, Cambridge, MA, USA.,Voyager Therapeutics, Cambridge, MA, USA
| | - L Chen
- Department of Neurology, Yale University, New Haven, CT, USA.,Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, USA
| | - M Alsaloum
- Department of Neurology, Yale University, New Haven, CT, USA.,Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, USA.,Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT, USA.,Yale Medical Scientist Training Program, Yale School of Medicine, New Haven, CT, USA
| | - N Rampal
- Neuroscience Department, Amgen Research, Thousand Oaks, CA, USA
| | - D J Matson
- Neuroscience Department, Amgen Research, Cambridge, MA, USA
| | - C Yang
- Neuroscience Department, Amgen Research, Cambridge, MA, USA
| | - K Taborn
- Neuroscience Department, Amgen Research, Cambridge, MA, USA.,Wave Life Sciences, Ltd, Cambridge, MA, USA
| | - M Zhang
- Neuroscience Department, Amgen Research, Thousand Oaks, CA, USA
| | - B Youngblood
- Neuroscience Department, Amgen Research, Thousand Oaks, CA, USA
| | - D Liu
- Neuroscience Department, Amgen Research, Thousand Oaks, CA, USA
| | - E Galbreath
- Comparative Biology and Safety Sciences, Amgen Research, Cambridge, MA, USA.,Takeda Pharmaceutical Company Ltd, Cambridge, MA, USA
| | - S Allred
- Comparative Biology and Safety Sciences, Amgen Research, South San Francisco, CA, USA.,Seattle Genetics, Bothell, WA, USA
| | - M Lepherd
- Comparative Biology and Safety Sciences, Amgen Research, South San Francisco, CA, USA.,Genentech, Inc. South San Francisco, CA, USA
| | - R Ferrando
- Comparative Biology and Safety Sciences, Amgen Research, South San Francisco, CA, USA.,AbbVie Stemcentrx, Inc., South San Francisco, CA, USA
| | - T J Kornecook
- Neuroscience Department, Amgen Research, Thousand Oaks, CA, USA.,Biogen Inc., Cambridge, MA, USA
| | - S G Lehto
- Neuroscience Department, Amgen Research, Thousand Oaks, CA, USA
| | - S G Waxman
- Department of Neurology, Yale University, New Haven, CT, USA.,Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, USA
| | - B D Moyer
- Neuroscience Department, Amgen Research, Thousand Oaks, CA, USA
| | - S Dib-Hajj
- Department of Neurology, Yale University, New Haven, CT, USA.,Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, USA
| | - J Gingras
- Neuroscience Department, Amgen Research, Cambridge, MA, USA.,Homology Medicine Inc., Bedford, MA, USA
| |
Collapse
|
27
|
Kushnarev M, Pirvulescu IP, Candido KD, Knezevic NN. Neuropathic pain: preclinical and early clinical progress with voltage-gated sodium channel blockers. Expert Opin Investig Drugs 2020; 29:259-271. [PMID: 32070160 DOI: 10.1080/13543784.2020.1728254] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Introduction: Neuropathic pain is a chronic condition that significantly affects the quality of life of millions of people globally. Most of the pharmacologic treatments currently in use demonstrate modest efficacy and over half of all patients do not respond to medical management. Hence, there is a need for new, efficacious drugs. Evidence points toward voltage-gated sodium channels as a key target for novel analgesics.Area covered: The role of voltage-gated sodium channels in pain pathophysiology is illuminated and the preclinical and clinical data for new sodium channel blockers and toxin-derived lead compounds are examined. The expansion of approved sodium channel blockers is discussed along with the limitations of current research, trends in drug development, and the potential of personalized medicine.Expert opinion: The transition from preclinical to clinical studies can be difficult because of the inherent inability of animal models to express the complexities of pain states. Pain pathways are notoriously intricate and may be pharmacologically modulated at a variety of targets; it is unlikely that action at a single target could completely abolish a pain response because pain is rarely unifactorial. Combination therapy may be necessary and this could further confound the discovery of novel agents.
Collapse
Affiliation(s)
- Mikhail Kushnarev
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, Chicago, IL, USA
| | - Iulia Paula Pirvulescu
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, Chicago, IL, USA
| | - Kenneth D Candido
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, Chicago, IL, USA.,Department of Anesthesiology, College of Medicine, University of Illinois, Chicago, IL, USA.,Department of Surgery, College of Medicine, University of Illinois, Chicago, IL, USA
| | - Nebojsa Nick Knezevic
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, Chicago, IL, USA.,Department of Anesthesiology, College of Medicine, University of Illinois, Chicago, IL, USA.,Department of Surgery, College of Medicine, University of Illinois, Chicago, IL, USA
| |
Collapse
|