Elsevier

Brain Research

Volume 1215, 18 June 2008, Pages 69-75
Brain Research

Research Report
The cannabinoid receptor agonist, WIN 55, 212-2, attenuates tumor-evoked hyperalgesia through peripheral mechanisms

https://doi.org/10.1016/j.brainres.2008.03.063Get rights and content

Abstract

Several lines of evidence suggest that cannabinoids can attenuate various types of pain and hyperalgesia through peripheral mechanisms. The development of rodent cancer pain models has provided the opportunity to investigate novel approaches to treat this common form of pain. In the present study, we examined the ability of peripherally administered cannabinoids to attenuate tumor-evoked mechanical hyperalgesia in a murine model of cancer pain. Unilateral injection of osteolytic fibrosarcoma cells into and around the calcaneus bone resulted in tumor formation and mechanical hyperalgesia in the injected hindpaw. Mechanical hyperalgesia was defined as an increase in the frequency of paw withdrawals to a suprathreshold von Frey filament (3.4 mN) applied to the plantar surface of the hindpaw. WIN 55, 212-2 (1.5 to 10 μg) injected subcutaneously into the tumor-bearing hindpaw produced a dose-dependent decrease in paw withdrawal frequencies to suprathreshold von Frey filament stimulation. Injection of WIN 55,212-2 (10 μg) into the contralateral hindpaw did not decrease paw withdrawal frequencies in the tumor-bearing hindpaw. Injection of the highest antihyperalgesic dose of WIN 55,212-2 (10 μg) did not produce catalepsy as determined by the bar test. Co-administration of WIN 55,212-2 with either cannabinoid 1 (AM251) or cannabinoid 2 (AM630) receptor antagonists attenuated the antihyperalgesic effects of WIN 55, 212-2. In conclusion, peripherally administered WIN 55,212-2 attenuated tumor-evoked mechanical hyperalgesia by activation of both peripheral cannabinoid 1 and cannabinoid 2 receptors. These results suggest that peripherally-administered cannabinoids may be effective in attenuating cancer pain.

Introduction

Pain resulting from cancer still presents a major therapeutic challenge. Current estimates indicate that over half of patients with cancer experience pain, and nearly two-thirds in advanced disease stages experience pain (van den Beuken-van Everdingen et al., 2007). Of those reporting cancer pain, it is estimated that one-third of all cancer patients rate their pain as moderate to severe (van den Beuken-van Everdingen et al., 2007). Although the prevalence of cancer pain differs among the various types of cancer, it is often reported by individuals with both primary and metastatic bone cancer, with greater than 60% of individuals with metastatic bone cancer suffering from severe pain (Coleman, 2006).

Cancer pain is most often treated with opioid drugs; however, these drugs produce dose-limiting side effects and sometimes provide inadequate analgesia (Hanks and Forbes, 1997, Cherny, 2004). Current estimates suggest that adequate analgesia is not achieved in all cancer patients using the World Health Organization's (WHO's) analgesic ladder (Azevedo et al., 2006). Understanding the mechanisms related to the generation and maintenance of cancer pain is needed for the development of novel and more efficacious therapies to treat this condition.

The use of cannabinoids to treat cancer pain may provide a novel therapeutic approach. Two endogenous receptors for cannabinoids have been isolated and cloned so far, cannabinoid 1 (CB1) and cannabinoid 2 (CB2) receptors (Matsuda et al., 1990, Murro et al., 1993), and both have been localized to various neuronal and non-neuronal tissues. Previous studies have shown that systemic administration of cannabinoids produces antinociception and attenuates hyperalgesia and allodynia in animal models of acute and chronic pain (for reviews see Walker et al., 1999, Hohmann, 2002, Walker and Huang, 2002, Mbvudula et al., 2004). The efficacy of systemically delivered cannabinoids is limited by adverse cannabimimetic effects (catalepsy, hypolocomotion, and disruptions in memory) which are mediated through CB1 receptor activation in the central nervous system (for review see Iversen, 2003). One way to avoid these centrally-mediated cannabimimetic effects, while still providing analgesia, is through the selective targeting of peripheral CB1 receptors expressed on nociceptive primary afferent dorsal root ganglion (DRG) neurons (Hohman and Hekenham, 1999; Ahluwalia et al., 2000) and cutaneous nerve terminals (Ständer et al., 2005, Amaya et al., 2006, Agarwal et al., 2007).

A recent study from our lab demonstrated that systemic administration of the non-selective cannabinoid receptor agonist CP 55,940 attenuated tumor-evoked mechanical hyperalgesia in an established murine model of cancer pain through activation of CB1 receptors (Hamamoto et al., 2007). In this cancer pain model, osteolytic fibrosarcoma cells were injected into and around the calcaneus bone which produces a tumor mass and eventually leads to hyperalgesia and sensitization of C nociceptors overlying the tumor on the plantar surface of the hindpaw (Cain et al., 2001, Wacnik et al., 2001). Since systemically administered cannabinoids can produce adverse cannabimimetic effects, the present study was designed to determine if local injection of the non-selective cannabinoid receptor agonist WIN 55,212-2 can attenuate tumor-evoked mechanical hyperalgesia and whether this effect occurred through activation of CB1 or CB2 receptors.

Section snippets

Tumor-evoked mechanical hyperalgesia

Prior to implantation of fibrosarcoma cells, the paw withdrawal frequency evoked by a 3.4 mN von Frey filament was 0–20% (data not shown). Mechanical hyperalgesia was fully developed by 6 days post-implantation at which time paw withdrawal frequencies increased in the tumor-bearing hindpaw to 70–100% (data not shown). No changes in paw withdrawal frequencies were observed in the contralateral hindpaw where paw withdrawal frequencies remained 0–20% (data not shown). These data are consistent

Discussion

In summary, local intraplantar injection of WIN 55,212-2 into the tumor-bearing hindpaw dose-dependently attenuated tumor-evoked mechanical hyperalgesia. The antihyperalgesic effects of WIN 55,212-2 were not the result of catalepsy and were blocked by co-administration of either CB1 or CB2 receptor antagonists. Injection of WIN 55,212-2 into the contralateral hindpaw did not alter mechanical hyperalgesia in the tumor-bearing hindpaw. Overall, these results show that local injection of WIN

Subjects

Adult (> 6 weeks old) male C3H/He mice weighing 25–29 g were used for this study. Animals were obtained from the National Institutes of Health (Frederick, MD), housed on a 12-hour light/dark schedule, and allowed ad libitum access to food and water. Each animal was used in only one experiment. All animal procedures and protocols were approved by the Animal Care Committee at the University of Minnesota, and experiments were conducted according to the guidelines established by the International

Acknowledgments

This work was supported by grants from the National Institutes of Health (CA91007 and DA011471 to D.A.S) and the U.S. Civilian Research and Development Foundation. C.P was supported by training grant from the National Institute on Drug Abuse (5T32-DA007234). The authors thank Dr. Thaddeus S. Brink for critically reading an early version of this paper.

References (52)

  • JohanekL.M. et al.

    Activation of peripheral cannabinoid receptors attenuates cutaneous hyperalgesia produced by a heat injury

    Pain

    (2004)
  • KehlL.J. et al.

    A cannabinoid agonist differentially attenuates deep tissue hyperalgesia in animal models of cancer and inflammatory muscle pain

    Pain

    (2003)
  • KhasabovaI. et al.

    Cannabinoids attenuate depolarization-dependent calcium influx in intermediate-size primary afferent neurons of adult rats

    Neuroscience

    (2002)
  • MenéndezL. et al.

    Analgesic effects of loperamide in bone cancer pain in mice

    Pharmacol. Biochem. Behav.

    (2005)
  • NackleyA.G. et al.

    Selective activation of cannabinoid CB(2) receptors suppresses spinal fos protein expression and pain behavior in a rat model of inflammation

    Neuroscience

    (2003)
  • RichardsonJ.D. et al.

    Cannabinoids reduce hyperalgesia and inflammation via interaction with peripheral CB1 receptors

    Pain

    (1998)
  • RossR.A. et al.

    Actions of cannabinoid receptor ligands on rat cultured sensory neurons: implications for antinociception

    Neuropharmacology

    (2001)
  • SalimK. et al.

    Pain measurements and side effect profile of the novel cannabinoid ajulemic acid

    Neuropharmacology

    (2005)
  • StänderS. et al.

    Distribution of cannabinoid receptor 1 (CB1) and 2 (CB2) on sensory nerve fibers and adnexal structures in human skin

    J. Dermatol. Sci.

    (2005)
  • van den Beuken-van EverdingenM.H. et al.

    Prevalence of pain in patients with cancer: a systematic review of the past 40 years

    Ann. Oncol.

    (2007)
  • WalkerJ.M. et al.

    Cannabinoid analgesia

    Pharmacol. Ther.

    (2002)
  • AgarwalN. et al.

    Cannabinoids mediate analgesia largely via peripheral type 1 cannabinoid receptors in nociceptors

    Nat. Neurosci.

    (2007)
  • Azevedo São Leão FerreiraK. et al.

    The WHO analgesic ladder for cancer pain control, twenty years of use How much pain relief does one get from using it?

    Support. Care Cancer

    (2006)
  • BaamondeA. et al.

    Effects of the local administration of selective mu-, delta-and kappa-opioid receptor agonists on osteosarcoma-induced hyperalgesia

    Naunyn-Schmiedeberg's Arch. Pharmacol.

    (2005)
  • CainD.M. et al.

    Functional interactions between tumor and peripheral nerve: changes in excitability and morphology of primary afferent fibers in a murine model of cancer pain

    J. Neurosci.

    (2001)
  • ChernyN.I.

    The pharmacologic management of cancer pain

    Oncology (Williston Park)

    (2004)
  • Cited by (42)

    • Involvement of spinal G-protein inwardly rectifying potassium (GIRK) channels in the enhanced antinociceptive effects of the activation of both μ-opioid and cannabinoid CB<inf>1</inf> receptors

      2022, Journal of Pharmacological Sciences
      Citation Excerpt :

      Previous studies have demonstrated that CP55,940 produces a catalepsy in rodents.23,24 Therefore, in case of animal studies, we mainly used WIN-55,212-2, which hardly causes the catalepsy in rodents in the following behavioral tests.25 Antinociceptive effects were evaluated by a hot-plate test (55 ± 0.5 °C; Muromachi Kikai Co., Ltd., Tokyo, Japan) as described previously.26

    View all citing articles on Scopus
    View full text