Elsevier

Neuropharmacology

Volume 59, Issue 3, September 2010, Pages 201-207
Neuropharmacology

A-887826 is a structurally novel, potent and voltage-dependent Nav1.8 sodium channel blocker that attenuates neuropathic tactile allodynia in rats

https://doi.org/10.1016/j.neuropharm.2010.05.009Get rights and content

Abstract

Activation of sodium channels is essential to action potential generation and propagation. Recent genetic and pharmacological evidence indicates that activation of Nav1.8 channels contributes to chronic pain. Herein, we describe the identification of a novel series of structurally related pyridine derivatives as potent Nav1.8 channel blockers. A-887826 exemplifies this series and potently (IC50 = 11nM) blocked recombinant human Nav1.8 channels. A-887826 was ∼3 fold less potent to block Nav1.2, ∼10 fold less potent to block tetrodotoxin-sensitive sodium (TTX-S Na+) currents and was >30 fold less potent to block NaV1.5 channels. A-887826 potently blocked tetrodotoxin-resistant sodium (TTX-R Na+) currents (IC50 = 8nM) from small diameter rat dorsal root ganglion (DRG) neurons in a voltage-dependent fashion. A-887826 effectively suppressed evoked action potential firing when DRG neurons were held at depolarized potentials and reversibly suppressed spontaneous firing in small diameter DRG neurons from complete Freund’s adjuvant inflamed rats. Following oral administration, A-887826 significantly attenuated tactile allodynia in a rat neuropathic pain model. Further characterization of TTX-R current block in rat DRG neurons demonstrated that A-887826 (100 nM) shifted the mid-point of voltage-dependent inactivation of TTX-R currents by ∼4 mV without affecting voltage-dependent activation and did not exhibit frequency-dependent inhibition. The present data demonstrate that A-887826 is a structurally novel and potent Nav1.8 blocker that inhibits rat DRG TTX-R currents in a voltage-, but not frequency-dependent fashion. The ability of this structurally novel Nav1.8 blocker to effectively reduce tactile allodynia in neuropathic rats further supports the role of Nav1.8 sodium channels in pathological pain states.

Introduction

Voltage-gated sodium channels play important roles in action potential generation and propagation (Waxman et al., 1999). At least 9 genes have been identified that encode functional sodium channels, namely Nav1.1–Nav1.9, and each subtype can be functionally classified as either tetrodotoxin-sensitive (TTX-S) or TTX-resistant (TTX-R) (Catterall et al., 2005). Both TTX-S and TTX-R sodium channels are expressed on primary afferent sensory neurons (Black et al., 2008, Renganathan et al., 2002). Recent pharmacological and genetic data have provided evidence for specific involvement of Nav1.3, 1.7, 1.8, and 1.9 in the processing of nociceptive information (Dib-Hajj et al., 2009). Of these, Nav1.8 is a TTX-R sodium channel that is highly localized on primary sensory afferent neurons (Akopian et al., 1999, Djouhri et al., 2003).

Studies using gene ablation, antisense (Porreca et al., 1999, Joshi et al., 2006) or small interfering RNA (siRNA) (Dong et al., 2007) to knockdown expression of Nav1.8 have demonstrated a significant reduction in mechanical allodynia and hyperalgesia in inflammatory and neuropathic pain models in rats. Suppression of Nav1.8 expression also produces a reduction in visceral pain in several experimental models (Yoshimura et al., 2001, Laird et al., 2002). The importance of Nav1.8 channels in pain sensitivity is also supported by recent data showing that activation of Nav1.8 sodium channels is the primary driver of nociceptor excitability under cold conditions (Zimmermann et al., 2007).

Systemic or spinal delivery of non-selective sodium channel blockers reduces hyperalgesia and allodynia in animal pain models (Veneroni et al., 2003, Brochu et al., 2006, Gaida et al., 2005, Akada et al., 2006, Ekberg et al., 2006). Previously, we reported the discovery of A-803467, a potent, voltage-dependent, and highly selective Nav1.8 channel blocker that effectively reduces chronic pain in preclinical models. The discovery of A-803467 demonstrated that pharmacological selective sodium channel blockers is achievable and also provided support for the role of Nav1.8 in chronic pain states (Jarvis et al., 2007).

In the present study, we report the discovery and characterization of A-887826, 5-(4-butoxy-3-chlorophenyl)-N-((2-morpholinopyridin-3-yl)methyl)nicotinamide (Fig. 1A). A-887826 and related pyridine analogs represent structurally novel and highly potent Nav1.8 sodium channel blockers that are chemically distinct from A-803467. A-887826 potently blocks Nav1.8 channels in a voltage-, but not frequency-, dependent fashion and is less active at other sodium channels including Nav1.2 and Nav1.5. Following systemic administration, A-887826 produced dose-related attenuation of tactile allodynia in an animal model of neuropathic pain.

Section snippets

Animals

Male Sprague–Dawley rats (Charles River, Wilmington, MA) weighing 200–300 grams were utilized for in vivo studies. All procedures involving animal use were carried out in accordance with the Abbott Laboratories Institutional Animal Care and Use Committee. Rats were maintained in a temperature-controlled facility with a 12 h light/dark cycle and had ad libitum access to water and regular rodent chow.

HEK cells expressing human sodium channels

Human embryonic kidney (HEK-293) cells expressing recombinant sodium channels were grown in

Pyridine-based blockers of recombinant human NaV1.8 Na+ currents

Based on the A-803467 furfuramide scaffold (Kort et al., 2008), a series of structurally related 5-aryl nicotinamide derivatives was prepared and evaluated for their ability to block recombinant human Nav1.8 channels (Table 1). Substitution at the 4-position provided potent Nav1.8 blocking activity (e.g., compound 1) and incorporation of an adjacent halogen (e.g., 3-chloro, compound 2) was well tolerated. The potency could be further enhanced by increasing steric bulk (e.g. compounds 3–6).

Discussion

The present data demonstrate that A-887826 potently blocks both recombinant human and native rat Nav1.8 sodium channels in a voltage-dependent fashion. A-887826 was more potent in blocking human Nav1.8 channels and native rat TTX-R Na+ currents compared to its activity at human Nav1.2 and Nav1.5 sodium channels and rat TTX-S Na+ currents. Additionally, A-887826 showed little or no activity in assays of a large array of other channels and receptors. A-887826 represents a structurally novel Nav

References (38)

  • N. Yoshimura et al.

    Suppression of the tetrodotoxin-resistant sodium channel (PN3/SNS): a possible new treatment for bladder pain

    Urology

    (2001)
  • X.F. Zhang et al.

    Differential action potentials and firing patterns in injured and uninjured small dorsal root ganglion neurons after nerve injury

    Brain Res.

    (2004)
  • R. Amir et al.

    The role of sodium channels in chronic inflammatory and neuropathic pain

    J. Pain

    (2006)
  • A.N. Akopian et al.

    The tetrodotoxin-resistant sodium channel SNS has a specialized function in pain pathways

    Nat. Neurosci.

    (1999)
  • J.A. Black et al.

    Multiple sodium channel isoforms and mitogen-activated protein kinases are present in painful human neuromas

    Ann. Neurol.

    (2008)
  • M.E. Bräu et al.

    Effect of drugs used for neuropathic pain management on tetrodotoxin-resistant Na(+) currents in rat sensory neurons

    Anesthesiology

    (2001)
  • R.M. Brochu et al.

    Block of peripheral nerve sodium channels selectively inhibits features of neuropathic pain in rats

    Mol. Pharmacol.

    (2006)
  • W.A. Catterall et al.

    International Union of Pharmacology. XLVII. Nomenclature and structure-function relationships of voltage-gated sodium channels

    Pharmacol. Rev.

    (2005)
  • S.D. Dib-Hajj et al.

    Voltage-gated sodium channels: therapeutic targets for pain

    Pain Med.

    (2009)
  • Cited by (43)

    • Voltage-Gated Ion Channels in the PNS: Novel Therapies for Neuropathic Pain?

      2016, Trends in Pharmacological Sciences
      Citation Excerpt :

      NaV1.8, too, has received significant attention with regard to drug development [173]. As with NaV1.7, both synthetic [174–181] and naturally occurring [182–184] small molecule NaV1.8-selective inhibitors have been described, which possess antihyperalgesic activity in preclinical models of neuropathic pain. Of those, PF-01247324 [6-amino-N-methyl-5-(2,3,5-trichlorophenyl)pyridine-2-carboxamide] appears particularly promising.

    • Voltage gated sodium and calcium channel blockers for the treatment of chronic inflammatory pain

      2013, Neuroscience Letters
      Citation Excerpt :

      These findings led to the development of a small molecule selective blocker of NaV1.8, A803467, which significantly attenuated hypersensitive behavior in a variety of animal models of inflammatory pain [67] including OA pain [105]. Improvements in the bioavailability of small molecule NaV1.8 blockers holds promise for their analgesic potential in treating chronic inflammatory pain states [103,140]. Interestingly, neurogenic inflammation and subsequent sensitization of dural afferents is thought to trigger migraine pain.

    • XInjury-Induced HDAC5 nuclear export is essential for axon regeneration

      2013, Cell
      Citation Excerpt :

      In contrast, blocking voltage-gated sodium channels (VGSCs) with TTX had little effect on calcium-wave back-propagation and calcium levels in cell body compared to control (Figures 1A and 1B; Movie S3). Because DRG neurons contain TTX-resistant sodium channels, we used A887826 (Zhang et al., 2010) to block Nav1.8, one of the most prominent TTX-resistant sodium channels present in the majority of DRG neurons (Shields et al., 2012). A887826, similar to TTX, did not significantly block calcium back-propagation and accumulation in the soma (Figures 1A and 1B; Movie S4).

    View all citing articles on Scopus
    View full text