HOMEPRODUCTSCOMPANYCONTACTFAQResearchDictionaryPharmaSign Up FREE or Login

Brain permeant and impermeant inhibitors of fatty-acid amide hydrolase suppress the development and maintenance of paclitaxel-induced neuropathic pain without producing tolerance or physical dependence in vivo and synergize with paclitaxel to reduce tumor cell line viability in vitro.

Abstract
Activation of cannabinoid CB1 receptors suppresses pathological pain but also produces unwanted side effects, including tolerance and physical dependence. Inhibition of fatty-acid amide hydrolase (FAAH), the major enzyme catalyzing the degradation of anandamide (AEA), an endocannabinoid, and other fatty-acid amides, suppresses pain without unwanted side effects typical of direct CB1 agonists. However, FAAH inhibitors have failed to show efficacy in several clinical trials suggesting that the right partnership of FAAH inhibition and pathology has yet to be identified. We compared efficacy of chronic treatments with a centrally penetrant FAAH inhibitor (URB597), a peripherally restricted FAAH inhibitor (URB937) and an orthosteric pan-cannabinoid agonist (WIN55,212-2) in suppressing neuropathic pain induced by the chemotherapeutic agent paclitaxel. Each FAAH inhibitor suppressed the development of paclitaxel-induced neuropathic pain and reduced the maintenance of already established allodynia with sustained efficacy. Tolerance developed to the anti-allodynic efficacy of WIN55,212-2, but not to that of URB597 or URB937, in each dosing paradigm. Challenge with the CB1 antagonist rimonabant precipitated CB1-dependent withdrawal in paclitaxel-treated mice receiving WIN55,212-2 but not URB597 or URB937. When dosing with either URB597 or URB937 was restricted to the development of neuropathy, paclitaxel-induced allodynia emerged following termination of drug delivery. These observations suggest that both FAAH inhibitors were anti-allodynic rather than curative. Moreover, neither URB597 nor URB937 impeded the ability of paclitaxel to reduce breast (4T1) or ovarian (HeyA8) tumor cell line viability. In fact, URB597 and URB937 alone reduced 4T1 tumor cell line viability, albeit with low potency, and the dose matrix of each combination with paclitaxel was synergistic in reducing 4T1 and HeyA8 tumor cell line viability according to Bliss, Highest Single Agent (HSA) and Loewe additivity models. Both FAAH inhibitors synergized with paclitaxel to reduce 4T1 and HeyA8 tumor cell line viability without reducing viability of non-tumor HEK293 cells. Neither FAAH inhibitor reduced viability of non-tumor HEK293 cells in either the presence or absence of paclitaxel, suggesting that nonspecific cytotoxic effects were not produced by the same treatments. Our results suggest that FAAH inhibitors reduce paclitaxel-induced allodynia without the occurrence of CB1-dependence in vivo and may, in fact, enhance the anti-tumor actions of paclitaxel in vitro.
AuthorsRichard A Slivicki, Zhili Xu, Sonali S Mali, Andrea G Hohmann
JournalPharmacological research (Pharmacol Res) Vol. 142 Pg. 267-282 (04 2019) ISSN: 1096-1186 [Electronic] Netherlands
PMID30739035 (Publication Type: Journal Article, Research Support, N.I.H., Extramural)
CopyrightCopyright © 2019. Published by Elsevier Ltd.
Chemical References
  • Analgesics
  • Antineoplastic Agents
  • Benzamides
  • Benzoxazines
  • Cannabinoids
  • Carbamates
  • Morpholines
  • Naphthalenes
  • URB937
  • cyclohexyl carbamic acid 3'-carbamoylbiphenyl-3-yl ester
  • (3R)-((2,3-dihydro-5-methyl-3-((4-morpholinyl)methyl)pyrrolo-(1,2,3-de)-1,4-benzoxazin-6-yl)(1-naphthalenyl))methanone
  • Amidohydrolases
  • fatty-acid amide hydrolase
  • Paclitaxel
Topics
  • Amidohydrolases (antagonists & inhibitors)
  • Analgesics (therapeutic use)
  • Animals
  • Antineoplastic Agents
  • Benzamides (therapeutic use)
  • Benzoxazines (therapeutic use)
  • Brain (metabolism)
  • Cannabinoids (therapeutic use)
  • Carbamates (therapeutic use)
  • Cell Line, Tumor
  • Cell Survival (drug effects)
  • Drug Synergism
  • Drug Tolerance
  • HEK293 Cells
  • Humans
  • Hyperalgesia (chemically induced, drug therapy)
  • Male
  • Mice
  • Mice, Inbred C57BL
  • Morpholines (therapeutic use)
  • Naphthalenes (therapeutic use)
  • Neuralgia (chemically induced, drug therapy)
  • Paclitaxel
  • Substance-Related Disorders

Join CureHunter, for free Research Interface BASIC access!

Take advantage of free CureHunter research engine access to explore the best drug and treatment options for any disease. Find out why thousands of doctors, pharma researchers and patient activists around the world use CureHunter every day.
Realize the full power of the drug-disease research graph!


Choose Username:
Email:
Password:
Verify Password:
Enter Code Shown: