Elsevier

Biological Psychiatry

Volume 77, Issue 5, 1 March 2015, Pages 475-487
Biological Psychiatry

Archival Report
Chronic Cannabinoid Receptor 2 Activation Reverses Paclitaxel Neuropathy Without Tolerance or Cannabinoid Receptor 1–Dependent Withdrawal

https://doi.org/10.1016/j.biopsych.2014.04.009Get rights and content

Abstract

Background

Mixed cannabinoid receptor 1 and 2 (CB1 and CB2) agonists such as Δ9-tetrahydrocannabinol (Δ9-THC) can produce tolerance, physical withdrawal, and unwanted CB1-mediated central nervous system side effects. Whether repeated systemic administration of a CB2-preferring agonist engages CB1 receptors or produces CB1-mediated side effects is unknown.

Methods

We evaluated antiallodynic efficacy, possible tolerance, and cannabimimetic side effects of repeated dosing with a CB2-preferring agonist AM1710 in a model of chemotherapy-induced neuropathy produced by paclitaxel using CB1 knockout (CB1KO), CB2 knockout (CB2KO), and wild-type (WT) mice. Comparisons were made with the prototypic classic cannabinoid Δ9-THC. We also explored the site and possible mechanism of action of AM1710.

Results

Paclitaxel-induced mechanical and cold allodynia developed to an equivalent degree in CB1KO, CB2KO, and WT mice. Both AM1710 and Δ9-THC suppressed established paclitaxel-induced allodynia in WT mice. In contrast to Δ9-THC, chronic administration of AM1710 did not engage CB1 activity or produce antinociceptive tolerance, CB1-mediated cannabinoid withdrawal, hypothermia, or motor dysfunction. Antiallodynic efficacy of systemic administration of AM1710 was absent in CB2KO mice and WT mice receiving the CB2 antagonist AM630, administered either systemically or intrathecally. Intrathecal administration of AM1710 also attenuated paclitaxel-induced allodynia in WT mice, but not CB2KO mice, implicating a possible role for spinal CB2 receptors in AM1710 antiallodynic efficacy. Finally, both acute and chronic administration of AM1710 decreased messenger RNA levels of tumor necrosis factor-α and monocyte chemoattractant protein 1 in lumbar spinal cord of paclitaxel-treated WT mice.

Conclusions

Our results highlight the potential of prolonged use of CB2 agonists for managing chemotherapy-induced allodynia with a favorable therapeutic ratio marked by sustained efficacy and absence of tolerance, physical withdrawal, or CB1-mediated side effects.

Section snippets

Subjects

Adult CB2KO mice, strain B6.129P2-CNR2(tm1Dgen/J) (Jackson Laboratory, Bar Harbor, Maine) and WT littermates (Jackson Laboratory) on C57BL/6J background, and CB1KO mice (generated as previously described (4)) and WT littermates (Charles River Laboratories, Wilmington, Massachusetts) on CD1 background, weighing 25–33 g and of both sexes, were used in these experiments. Mice were periodically backcrossed to maintain genetic integrity. Animals were single-housed in a temperature-controlled

Paclitaxel-Induced Allodynia Developed Similarly in WT, CB2KO, and CB1KO Mice

In both CB2KO and WT mice, paclitaxel decreased mechanical thresholds [F3,20 = 519.03, p < .0001] (Figure 1A) and increased response time to cold stimulation [F3,20 = 553.78, p < .0001] (Figure 1B). Similarly, paclitaxel induced mechanical [F3,20 = 426.66, p < .0001] (Figure 1C) and cold [F3,20 = 707.28, p < .0001] (Figure 1D) allodynia in CB1KO mice and WT littermates. Mechanical and cold allodynia were present in paclitaxel-treated CB2KO, CB1KO, and WT mice relative to cremophor-vehicle since

Discussion

Drug development for management of neuropathic pain has been a challenge partly because of limited efficacy and troubling side-effect profiles. These challenges also apply to potential therapeutic use of cannabinoids (44). In the present study, we showed that repeated systemic administration of the CB2-preferring agonist AM1710 suppressed chemotherapy-induced allodynia without tolerance or significant CB1 involvement (i.e., the absence of CB1 antagonist–precipitated withdrawal symptoms, motor

Acknowledgments and Disclosures

This work was supported by National Institute on Drug Abuse (NIDA) Grant Nos. DA021644 (AGH), DA037673 (AGH), DA011322 (KM), DA021696 (KM), DA3801 (AM), DA07215 (AM), DA09158 (AM), and DA035068 (KM and AGH). AM serves as a consultant for MAKScientific.

We thank Vishnu Kodumuru for providing AM1710 and James Wager-Miller for designing and providing the reverse transcription polymerase chain reaction primers.

The authors report no biomedical financial interests or potential conflicts of interest.

References (91)

  • C. La Porta et al.

    Role of CB1 and CB2 cannabinoid receptors in the development of joint pain induced by monosodium iodoacetate

    Pain

    (2013)
  • G. Wotherspoon et al.

    Peripheral nerve injury induces cannabinoid receptor 2 protein expression in rat sensory neurons

    Neuroscience

    (2005)
  • L. Luongo et al.

    1-(2′,4′-dichlorophenyl)-6-methyl-N-cyclohexylamine-1,4-dihydroindeno[1,2-c]pyraz ole-3-carboxamide, a novel CB2 agonist, alleviates neuropathic pain through functional microglial changes in mice

    Neurobiol Dis

    (2010)
  • E. Burgos et al.

    Cannabinoid agonist WIN 55,212-2 prevents the development of paclitaxel-induced peripheral neuropathy in rats. Possible involvement of spinal glial cells

    Eur J Pharmacol

    (2012)
  • K.J. Valenzano et al.

    Pharmacological and pharmacokinetic characterization of the cannabinoid receptor 2 agonist, GW405833, utilizing rodent models of acute and chronic pain, anxiety, ataxia and catalepsy

    Neuropharmacology

    (2005)
  • H. McQuay

    Opioids in pain management

    Lancet

    (1999)
  • S. Gonzalez et al.

    Cannabinoid tolerance and dependence: A review of studies in laboratory animals

    Pharmacol Biochem Behav

    (2005)
  • S.P. Welch

    Characterization of anandamide-induced tolerance: Comparison to delta 9-THC-induced interactions with dynorphinergic systems

    Drug Alcohol Depend

    (1997)
  • C.E. Bass et al.

    Time course for the induction and maintenance of tolerance to Delta(9)-tetrahydrocannabinol in mice

    Drug Alcohol Depend

    (2000)
  • E.J. Rahn et al.

    Pharmacological characterization of AM1710, a putative cannabinoid CB2 agonist from the cannabilactone class: Antinociception without central nervous system side-effects

    Pharmacol Biochem Behav

    (2011)
  • S.G. Kinsey et al.

    The CB2 cannabinoid receptor-selective agonist O-3223 reduces pain and inflammation without apparent cannabinoid behavioral effects

    Neuropharmacology

    (2011)
  • M. Ozek et al.

    Comparison of the effects of specific and nonspecific inhibition of nitric oxide synthase on morphine analgesia, tolerance and dependence in mice

    Life Sci

    (2003)
  • K. Tsou et al.

    Physical withdrawal in rats tolerant to delta 9-tetrahydrocannabinol precipitated by a cannabinoid receptor antagonist

    Eur J Pharmacol

    (1995)
  • T. Gutierrez et al.

    Self-medication of a cannabinoid CB2 agonist in an animal model of neuropathic pain

    Pain

    (2011)
  • H. Zhang et al.

    Evidence that spinal astrocytes but not microglia contribute to the pathogenesis of paclitaxel-induced painful neuropathy

    J Pain

    (2012)
  • H. Zhang et al.

    Induction of monocyte chemoattractant protein-1 (MCP-1) and its receptor CCR2 in primary sensory neurons contributes to paclitaxel-induced peripheral neuropathy

    J Pain

    (2013)
  • N. Kiguchi et al.

    Up-regulation of tumor necrosis factor-alpha in spinal cord contributes to vincristine-induced mechanical allodynia in mice

    Neurosci Lett

    (2008)
  • L. Zhang et al.

    TNF-alpha contributes to spinal cord synaptic plasticity and inflammatory pain: Distinct role of TNF receptor subtypes 1 and 2

    Pain

    (2011)
  • Y.L. Liu et al.

    Tumor necrosis factor-alpha induces long-term potentiation of C-fiber evoked field potentials in spinal dorsal horn in rats with nerve injury: The role of NF-kappa B, JNK and p38 MAPK

    Neuropharmacology

    (2007)
  • H. Junger et al.

    Nociceptive and inflammatory effects of subcutaneous TNFalpha

    Pain

    (2000)
  • P. Pacher et al.

    Modulating the endocannabinoid system in human health and disease—successes and failures

    FEBS J

    (2013)
  • R.G. Pertwee

    Targeting the endocannabinoid system with cannabinoid receptor agonists: Pharmacological strategies and therapeutic possibilities

    Philos Trans R Soc Lond B Biol Sci

    (2012)
  • G. Tanda et al.

    Cannabinoids: Reward, dependence, and underlying neurochemical mechanisms—a review of recent preclinical data

    Psychopharmacology

    (2003)
  • C. Ledent et al.

    Unresponsiveness to cannabinoids and reduced addictive effects of opiates in CB1 receptor knockout mice

    Science

    (1999)
  • B.K. Atwood et al.

    CB2: A cannabinoid receptor with an identity crisis

    Br J Pharmacol

    (2010)
  • J. Guindon et al.

    The endocannabinoid system and pain

    CNS Neurol Disord Drug Targets

    (2009)
  • J.C. Ashton et al.

    The cannabinoid CB2 receptor as a target for inflammation-dependent neurodegeneration

    Curr Neuropharmacol

    (2007)
  • O. Sagredo et al.

    Cannabinoid CB2 receptor agonists protect the striatum against malonate toxicity: Relevance for Huntington’s disease

    Glia

    (2009)
  • C. Benito et al.

    Cannabinoid CB2 receptors and fatty acid amide hydrolase are selectively overexpressed in neuritic plaque-associated glia in Alzheimer’s disease brains

    J Neurosci

    (2003)
  • M.D. Jhaveri et al.

    Cannabinoid CB2 receptor-mediated anti-nociception in models of acute and chronic pain

    Mol Neurobiol

    (2007)
  • T.P. Malan et al.

    CB2 cannabinoid receptor-mediated peripheral antinociception

    Pain

    (2001)
  • A. Quartilho et al.

    Inhibition of inflammatory hyperalgesia by activation of peripheral CB2 cannabinoid receptors

    Anesthesiology

    (2003)
  • G.C. Hsieh et al.

    Central and peripheral sites of action for CB(2) receptor mediated analgesic activity in chronic inflammatory and neuropathic pain models in rats

    Br J Pharmacol

    (2011)
  • A. Romero-Sandoval et al.

    Spinal microglial and perivascular cell cannabinoid receptor type 2 activation reduces behavioral hypersensitivity without tolerance after peripheral nerve injury

    Anesthesiology

    (2008)
  • V. Curto-Reyes et al.

    Spinal and peripheral analgesic effects of the CB2 cannabinoid receptor agonist AM1241 in two models of bone cancer-induced pain

    Br J Pharmacol

    (2010)
  • Cited by (152)

    • Mechanisms of cannabinoid tolerance

      2023, Biochemical Pharmacology
    View all citing articles on Scopus
    View full text