Your browser doesn't support javascript.
loading
Discovery of a selective, state-independent inhibitor of NaV1.7 by modification of guanidinium toxins.
Pajouhesh, H; Beckley, J T; Delwig, A; Hajare, H S; Luu, G; Monteleone, D; Zhou, X; Ligutti, J; Amagasu, S; Moyer, B D; Yeomans, D C; Du Bois, J; Mulcahy, J V.
Affiliation
  • Pajouhesh H; SiteOne Therapeutics, South San Francisco, CA, 94080, USA.
  • Beckley JT; SiteOne Therapeutics, Bozeman, MT, 59715, USA.
  • Delwig A; SiteOne Therapeutics, South San Francisco, CA, 94080, USA.
  • Hajare HS; Department of Chemistry, Stanford University, Stanford, CA, 94305, USA.
  • Luu G; SiteOne Therapeutics, South San Francisco, CA, 94080, USA.
  • Monteleone D; SiteOne Therapeutics, South San Francisco, CA, 94080, USA.
  • Zhou X; SiteOne Therapeutics, South San Francisco, CA, 94080, USA.
  • Ligutti J; Neuroscience Department, Amgen Research, Thousand Oaks, CA, 91320, USA.
  • Amagasu S; Neuroscience Department, Amgen Research, Thousand Oaks, CA, 91320, USA.
  • Moyer BD; Neuroscience Department, Amgen Research, Thousand Oaks, CA, 91320, USA.
  • Yeomans DC; Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University, Stanford, CA, 94305, USA.
  • Du Bois J; Department of Chemistry, Stanford University, Stanford, CA, 94305, USA.
  • Mulcahy JV; SiteOne Therapeutics, South San Francisco, CA, 94080, USA. john.mulcahy@site1therapeutics.com.
Sci Rep ; 10(1): 14791, 2020 09 09.
Article in En | MEDLINE | ID: mdl-32908170
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.
Subject(s)

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Guanidine / Sodium Channel Blockers / NAV1.7 Voltage-Gated Sodium Channel Type of study: Guideline / Prognostic_studies Limits: Animals / Humans Language: En Journal: Sci Rep Year: 2020 Document type: Article Affiliation country: United States

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Guanidine / Sodium Channel Blockers / NAV1.7 Voltage-Gated Sodium Channel Type of study: Guideline / Prognostic_studies Limits: Animals / Humans Language: En Journal: Sci Rep Year: 2020 Document type: Article Affiliation country: United States