Skip to main content
Top
Published in: CNS Drugs 8/2008

01-08-2008 | Review Article

Role of Cannabinoids in the Management of Neuropathic Pain

Authors: Dr M. Isabel Martín Fontelles, Carlos Goicoechea García

Published in: CNS Drugs | Issue 8/2008

Login to get access

Abstract

The treatment of pain, particularly neuropathic pain, is one of the therapeutic applications of cannabis and cannabinoids that is currently under investigation and that stimulates interest among clinicians and basic researchers.
Animal pain models, including models of acute, antinociceptive, inflammatory and neuropathic pain, have demonstrated the antinociceptive efficacy of cannabinoids without causing serious alterations in animal behaviour. These data, together with the historic and current empiric use of cannabinoids, support the interest in the analysis of their effectiveness in treating neuropathic pain.
The evaluation of controlled trials that focus on the effect of cannabinoids on neuropathic pain reveals that this class of drugs is able to significantly reduce pain perception. Nevertheless, this effect is generally weak and clinical relevance remains under evaluation. Moreover, there is a lack of controlled trials and, in particular, comparisons with other drugs generally used in the treatment of neuropathic pain.
Despite the fact that further research is required to achieve a definitive assessment, current data obtained from basic research and from analysis of the available controlled trials indicate that cannabinoids can be accepted as a useful option in the treatment of neuropathic pain.
Literature
1.
go back to reference Mechoulam R. The pharmacohistory of cannabis sativa. In: Mechoulam R, editor. Cannabinoids as therapeutic agents. Boca Raton (FL): CRC Press, 1986: 1–20 Mechoulam R. The pharmacohistory of cannabis sativa. In: Mechoulam R, editor. Cannabinoids as therapeutic agents. Boca Raton (FL): CRC Press, 1986: 1–20
2.
go back to reference Reynolds JR. Therapeutical uses and toxic effects of Cannabis indica [letter]. Lancet 1890; 22: 637CrossRef Reynolds JR. Therapeutical uses and toxic effects of Cannabis indica [letter]. Lancet 1890; 22: 637CrossRef
3.
go back to reference Gaoni Y, Mechoulam R. Isolation, structure and partial synthesis of an active constituent of hashish. J Am Chem Soc 1964; 86: 1646–7CrossRef Gaoni Y, Mechoulam R. Isolation, structure and partial synthesis of an active constituent of hashish. J Am Chem Soc 1964; 86: 1646–7CrossRef
4.
go back to reference Devane WA, Dysarz FA, Johnson MR, et al. Determination and characterization of a cannabinoid receptor in rat brain. Mol Pharmacol 1988; 34: 605–13PubMed Devane WA, Dysarz FA, Johnson MR, et al. Determination and characterization of a cannabinoid receptor in rat brain. Mol Pharmacol 1988; 34: 605–13PubMed
5.
go back to reference Matsuda LA, Lolait SJ, Brownstein MJ, et al. Structure of a cannabinoid receptor and functional expression of the cloned cDNA. Nature 1990; 346: 561–4PubMedCrossRef Matsuda LA, Lolait SJ, Brownstein MJ, et al. Structure of a cannabinoid receptor and functional expression of the cloned cDNA. Nature 1990; 346: 561–4PubMedCrossRef
6.
go back to reference Devane WA, Hanus L, Breuer A, et al. Isolation and structure of a brain constituent that binds to the cannabinoid receptor. Science 1992; 258: 1946–9PubMedCrossRef Devane WA, Hanus L, Breuer A, et al. Isolation and structure of a brain constituent that binds to the cannabinoid receptor. Science 1992; 258: 1946–9PubMedCrossRef
7.
go back to reference Pertwee RG. The therapeutic potential of drugs that target cannabinoid receptors or modulate the tissue levels or actions of endocannabinoids. AAPS J 2005; 7(3): E625–54PubMedCrossRef Pertwee RG. The therapeutic potential of drugs that target cannabinoid receptors or modulate the tissue levels or actions of endocannabinoids. AAPS J 2005; 7(3): E625–54PubMedCrossRef
8.
go back to reference Herkenham M, Lynn AB, Little MD, et al. Cannabinoid receptor localization in brain. Proc Natl Acad Sci U S A 1990; 87: 1932–6PubMedCrossRef Herkenham M, Lynn AB, Little MD, et al. Cannabinoid receptor localization in brain. Proc Natl Acad Sci U S A 1990; 87: 1932–6PubMedCrossRef
9.
go back to reference Pertwee RG. Pharmacology of cannabinoid CB1 and CB2 receptors. Pharmacol Ther 1997; 74: 129–80PubMed Pertwee RG. Pharmacology of cannabinoid CB1 and CB2 receptors. Pharmacol Ther 1997; 74: 129–80PubMed
10.
go back to reference Pertwee RG. Pharmacological, physiological and clinical implications of the discovery of cannabinoid receptors. Biochem Soc Trans 1998; 26(2): 267–72PubMed Pertwee RG. Pharmacological, physiological and clinical implications of the discovery of cannabinoid receptors. Biochem Soc Trans 1998; 26(2): 267–72PubMed
11.
go back to reference Skaper SD, Buriani A, Dal Toso R, et al. The ALIAmide palmitoylethanolamide and cannabinoids, but not anandamide, are protective in a delayed postglutamate paradigm of excitotoxic death in cerebellar granule neurons. Proc Nat Acad Sci U S A 1996; 93(9): 3984–9CrossRef Skaper SD, Buriani A, Dal Toso R, et al. The ALIAmide palmitoylethanolamide and cannabinoids, but not anandamide, are protective in a delayed postglutamate paradigm of excitotoxic death in cerebellar granule neurons. Proc Nat Acad Sci U S A 1996; 93(9): 3984–9CrossRef
12.
go back to reference Sagan S, Venance L, Torrens Y, et al. Anandamide and WIN 55212-2 inhibit cyclic AMP formation through G-proteincoupled receptors distinct from CB1 cannabinoid receptors in cultured astrocytes. Eur J Neurosci 1999; 11(2): 691–9PubMedCrossRef Sagan S, Venance L, Torrens Y, et al. Anandamide and WIN 55212-2 inhibit cyclic AMP formation through G-proteincoupled receptors distinct from CB1 cannabinoid receptors in cultured astrocytes. Eur J Neurosci 1999; 11(2): 691–9PubMedCrossRef
13.
go back to reference Griffin G, Fernando SR, Ross RA, et al. Evidence for the presence of CB2-like cannabinoid receptors on peripheral nerve terminals. Eur J Pharmacol 1997; 339: 53–61PubMedCrossRef Griffin G, Fernando SR, Ross RA, et al. Evidence for the presence of CB2-like cannabinoid receptors on peripheral nerve terminals. Eur J Pharmacol 1997; 339: 53–61PubMedCrossRef
14.
go back to reference Calignano A, La Rana G, Giuffrida A, et al. Control of pain initiation by endogenous cannabinoids. Nature 1998; 394(6690): 277–81PubMedCrossRef Calignano A, La Rana G, Giuffrida A, et al. Control of pain initiation by endogenous cannabinoids. Nature 1998; 394(6690): 277–81PubMedCrossRef
15.
go back to reference Malan P, Ibrahim M, Deng H, et al. CB2 cannabinoid receptormediated peripheral antinociception. Pain 2001; 93: 239–45PubMedCrossRef Malan P, Ibrahim M, Deng H, et al. CB2 cannabinoid receptormediated peripheral antinociception. Pain 2001; 93: 239–45PubMedCrossRef
16.
go back to reference Mechoulam R, Ben-Shabat S, Hanus L, et al. Identification of an endogenous 2-monoglyceride, present in canine gut, that binds to cannabinoid receptors. Biochem Pharmacol 1995; 50: 83–90PubMedCrossRef Mechoulam R, Ben-Shabat S, Hanus L, et al. Identification of an endogenous 2-monoglyceride, present in canine gut, that binds to cannabinoid receptors. Biochem Pharmacol 1995; 50: 83–90PubMedCrossRef
17.
go back to reference Pertwee R, Ross RA. Cannabinoid receptors and their ligands. Prostaglandins Leukot Essent Fatty Acids 2002; 66: 101–21PubMedCrossRef Pertwee R, Ross RA. Cannabinoid receptors and their ligands. Prostaglandins Leukot Essent Fatty Acids 2002; 66: 101–21PubMedCrossRef
18.
go back to reference Howlet AC, Barth F, Bonner T, et al. International Union of Pharmacology: XXVII. Classification of cannabinoid receptors. Pharmacol Rev 2002; 54: 161–202 Howlet AC, Barth F, Bonner T, et al. International Union of Pharmacology: XXVII. Classification of cannabinoid receptors. Pharmacol Rev 2002; 54: 161–202
19.
go back to reference Goya P, Jagerovic N, Hernández-Folgado L, et al. Cannabinoids and neuropathic pain. Mini Rev Med Chem 2003; 3(7): 765–72PubMedCrossRef Goya P, Jagerovic N, Hernández-Folgado L, et al. Cannabinoids and neuropathic pain. Mini Rev Med Chem 2003; 3(7): 765–72PubMedCrossRef
20.
go back to reference Hillard CJ, Jarrahian A. Cellular accumulation of anandamide: consensus and controversy. Br J Pharmacol 2003; 140: 802–8PubMedCrossRef Hillard CJ, Jarrahian A. Cellular accumulation of anandamide: consensus and controversy. Br J Pharmacol 2003; 140: 802–8PubMedCrossRef
21.
go back to reference Stella N, Schweitzer P, Piomelli D. A second endogenous cannabinoid that modulates long-term potentiation. Nature 1997; 388: 773–8PubMedCrossRef Stella N, Schweitzer P, Piomelli D. A second endogenous cannabinoid that modulates long-term potentiation. Nature 1997; 388: 773–8PubMedCrossRef
22.
go back to reference Hanus L, Abu Lafi SS, Fride E, et al. 2-Arachidonyl glyceryl ether, an endogenous agonist of the cannabinoid CB1 receptor. Proc Natl Acad Sci U S A 2001; 98: 3662–5PubMedCrossRef Hanus L, Abu Lafi SS, Fride E, et al. 2-Arachidonyl glyceryl ether, an endogenous agonist of the cannabinoid CB1 receptor. Proc Natl Acad Sci U S A 2001; 98: 3662–5PubMedCrossRef
23.
go back to reference Porter AC, Sauer JM, Knierman MD, et al. Characterization of a novel endocannabinoid, virodhamine, with antagonist activity at the CB1 receptor. J Pharmacol Exp Ther 2002; 301: 1020–4PubMedCrossRef Porter AC, Sauer JM, Knierman MD, et al. Characterization of a novel endocannabinoid, virodhamine, with antagonist activity at the CB1 receptor. J Pharmacol Exp Ther 2002; 301: 1020–4PubMedCrossRef
24.
go back to reference Huang SM, Bisogno T, Trevisani M, et al. An endogenous capsaicin-like substance with high potency at recombinant and native vanilloid VR1 receptors. Proc Natl Acad Sci U S A 2002; 99: 8400–5PubMedCrossRef Huang SM, Bisogno T, Trevisani M, et al. An endogenous capsaicin-like substance with high potency at recombinant and native vanilloid VR1 receptors. Proc Natl Acad Sci U S A 2002; 99: 8400–5PubMedCrossRef
25.
26.
go back to reference Di Marzo V, De Petrocellis L, Bisogno T. The biosynthesis, fate and pharmacological properties of endocannabinoids. In: Pertwee RG, editor. Cannabinoids: handbook of experimental pharmacology. Vol. 168. Heidelburg: Springer-Verlag, 2005: 147–85 Di Marzo V, De Petrocellis L, Bisogno T. The biosynthesis, fate and pharmacological properties of endocannabinoids. In: Pertwee RG, editor. Cannabinoids: handbook of experimental pharmacology. Vol. 168. Heidelburg: Springer-Verlag, 2005: 147–85
28.
go back to reference Buxbaum DM. Analgesic activity of delta-9-tetrahydrocannabinol in the rat and mouse. Psychopharmacologia 1972; 25: 275–80PubMedCrossRef Buxbaum DM. Analgesic activity of delta-9-tetrahydrocannabinol in the rat and mouse. Psychopharmacologia 1972; 25: 275–80PubMedCrossRef
29.
go back to reference Sofia RD, Vassar HB, Knobloch LC. Comparative analgesic activity of various naturally occurring cannabinoids in rats and mice. Psychopharmacologia 1975; 40: 285–95PubMedCrossRef Sofia RD, Vassar HB, Knobloch LC. Comparative analgesic activity of various naturally occurring cannabinoids in rats and mice. Psychopharmacologia 1975; 40: 285–95PubMedCrossRef
30.
go back to reference Johnson MR, Melvin LS, Milne GM. Prototype cannabinoid analgetics, prostaglandins and opiate search for points of mechanistic interaction. Life Sci 1982; 31: 1703–6PubMedCrossRef Johnson MR, Melvin LS, Milne GM. Prototype cannabinoid analgetics, prostaglandins and opiate search for points of mechanistic interaction. Life Sci 1982; 31: 1703–6PubMedCrossRef
31.
go back to reference Dajani EZ, Larsen KR, Taylor J, et al. 19,19-Dimethylheptyl-D-8-tetrahydrocannabinol-11-oic acid: a novel, orally effective cannabinoid with analgesic and anti-inflammatory properties. J Pharmacol Experiment Ther 1999; 291: 31–8 Dajani EZ, Larsen KR, Taylor J, et al. 19,19-Dimethylheptyl-D-8-tetrahydrocannabinol-11-oic acid: a novel, orally effective cannabinoid with analgesic and anti-inflammatory properties. J Pharmacol Experiment Ther 1999; 291: 31–8
33.
go back to reference De Vry J, Denzer D, Reissmueller E, et al. 3-[2-cyano-3-(trifluoromethyl)phenoxy]phenyl-4,4,4-trifluoro-l-butanesulfonate (BAY 59-3074): a novel cannabinoid Cbl/Cb2 receptor partial agonist with antihyperalgesic and antiallodynic effects. J Pharmacol Exp Ther 2004; 310(2): 620–32PubMedCrossRef De Vry J, Denzer D, Reissmueller E, et al. 3-[2-cyano-3-(trifluoromethyl)phenoxy]phenyl-4,4,4-trifluoro-l-butanesulfonate (BAY 59-3074): a novel cannabinoid Cbl/Cb2 receptor partial agonist with antihyperalgesic and antiallodynic effects. J Pharmacol Exp Ther 2004; 310(2): 620–32PubMedCrossRef
34.
go back to reference Smith FL, Fujimori K, Lowe J, et al. Characterization of delta9-tetrahydrocannabinol and anandamide antinociception in nonarthritic and arthritic rats. Pharm Biochem Behavior 1998; 60(1): 183–91CrossRef Smith FL, Fujimori K, Lowe J, et al. Characterization of delta9-tetrahydrocannabinol and anandamide antinociception in nonarthritic and arthritic rats. Pharm Biochem Behavior 1998; 60(1): 183–91CrossRef
35.
go back to reference Choong KC, Su X, Urban MO. Effect of CP55,940 on mechanosensory spinal neurons following chronic inflammation. Neurosci Lett 2007; 414(2): 105–9PubMedCrossRef Choong KC, Su X, Urban MO. Effect of CP55,940 on mechanosensory spinal neurons following chronic inflammation. Neurosci Lett 2007; 414(2): 105–9PubMedCrossRef
36.
go back to reference Vann RE, Cook CD, Martin BR, et al. Cannabimimetic properties of ajulemic acid. J Pharmacol Exp Ther 2007; 320(2): 678–86PubMedCrossRef Vann RE, Cook CD, Martin BR, et al. Cannabimimetic properties of ajulemic acid. J Pharmacol Exp Ther 2007; 320(2): 678–86PubMedCrossRef
37.
go back to reference Beaulieu P, Bisogno T, Punwar S, et al. Role of the endogenous cannabinoid system in the formalin test of persistent pain in the rat. Eur J Pharmacol 2000; 396(2–3): 85–92CrossRef Beaulieu P, Bisogno T, Punwar S, et al. Role of the endogenous cannabinoid system in the formalin test of persistent pain in the rat. Eur J Pharmacol 2000; 396(2–3): 85–92CrossRef
38.
go back to reference Guindon J, Desroches J, Beaulieu P. The antinociceptive effects of intraplantar injections of 2-arachidonoyl glycerol are mediated by cannabinoid CB2 receptors. Br J Pharmacol 2007; 150(6): 693–701PubMedCrossRef Guindon J, Desroches J, Beaulieu P. The antinociceptive effects of intraplantar injections of 2-arachidonoyl glycerol are mediated by cannabinoid CB2 receptors. Br J Pharmacol 2007; 150(6): 693–701PubMedCrossRef
39.
go back to reference Calignano A, La Rana G, Iufrida A, et al. Control of pain initiation by endogenous cannabinoids. Nature 1998; 396: 277–81 Calignano A, La Rana G, Iufrida A, et al. Control of pain initiation by endogenous cannabinoids. Nature 1998; 396: 277–81
40.
go back to reference Amaya F, Shimosato G, Kawasaki Y, et al. Induction of CB1 cannabinoid receptor by inflammation in primary afferent neurons facilitates antihyperalgesic effect of peripheral CB1 agonist. Pain 2006; 124(1-2): 175–83PubMedCrossRef Amaya F, Shimosato G, Kawasaki Y, et al. Induction of CB1 cannabinoid receptor by inflammation in primary afferent neurons facilitates antihyperalgesic effect of peripheral CB1 agonist. Pain 2006; 124(1-2): 175–83PubMedCrossRef
41.
go back to reference Romero-Sandoval A, Eisenach JC. Spinal cannabinoid receptor type 2 activation reduces hypersensitivity and spinal cord glial activation after paw incision. Anesthesiology 2007; 106(4): 787–94PubMedCrossRef Romero-Sandoval A, Eisenach JC. Spinal cannabinoid receptor type 2 activation reduces hypersensitivity and spinal cord glial activation after paw incision. Anesthesiology 2007; 106(4): 787–94PubMedCrossRef
42.
go back to reference Vann RE, Cook CD, Martin BR, et al. Cannabimimetic properties of ajulemic acid. J Pharmacol Exp Ther 2007; 320(2): 678–86PubMedCrossRef Vann RE, Cook CD, Martin BR, et al. Cannabimimetic properties of ajulemic acid. J Pharmacol Exp Ther 2007; 320(2): 678–86PubMedCrossRef
43.
go back to reference La Rana G, Russo R, Campolongo P, et al. Modulation of neuropathic and inflammatory pain by the endocannabinoid transport inhibitor AM404 [N-(4-hydroxyphenyl)-eicosa-5,8,11,14-tetraenamide]. J Pharmacol Exp Ther 2006; 317(3): 1365–71PubMedCrossRef La Rana G, Russo R, Campolongo P, et al. Modulation of neuropathic and inflammatory pain by the endocannabinoid transport inhibitor AM404 [N-(4-hydroxyphenyl)-eicosa-5,8,11,14-tetraenamide]. J Pharmacol Exp Ther 2006; 317(3): 1365–71PubMedCrossRef
44.
go back to reference Martin WJ, Loo CM, Basbaum AI. Spinal cannabinoids are antiallodynic in rats with persistent inflammation. Pain 1999; 82(2): 199–205PubMedCrossRef Martin WJ, Loo CM, Basbaum AI. Spinal cannabinoids are antiallodynic in rats with persistent inflammation. Pain 1999; 82(2): 199–205PubMedCrossRef
45.
go back to reference Agarwal N, Pacher P, Tegeder I, et al. Cannbinoids mediate analgesia largely via peripheral type cannabioid receptors in nociceptors. Nat Neurosci 2007; 10(7): 870–9PubMedCrossRef Agarwal N, Pacher P, Tegeder I, et al. Cannbinoids mediate analgesia largely via peripheral type cannabioid receptors in nociceptors. Nat Neurosci 2007; 10(7): 870–9PubMedCrossRef
46.
go back to reference Jeske NA, Patwardhan AM, Gamper N, et al. Cannabinoid WIN 55.212-2 regulates TRPV1 phosphorialtion in sensory neurons. J Biol Chem 2006; 281(43): 32879–90PubMedCrossRef Jeske NA, Patwardhan AM, Gamper N, et al. Cannabinoid WIN 55.212-2 regulates TRPV1 phosphorialtion in sensory neurons. J Biol Chem 2006; 281(43): 32879–90PubMedCrossRef
47.
go back to reference Costa B, Trovato AE, Comelli F, et al. The non-psychoactive cannabis constituent cannabidiol is an orally effective therapeutic agent in rat chronic inflammatory and neuropathic pain. Eur J Pharmacol 2007; 556(1–3): 75–83PubMedCrossRef Costa B, Trovato AE, Comelli F, et al. The non-psychoactive cannabis constituent cannabidiol is an orally effective therapeutic agent in rat chronic inflammatory and neuropathic pain. Eur J Pharmacol 2007; 556(1–3): 75–83PubMedCrossRef
48.
go back to reference Besse D, Lombard MC, Besson JM. Time-related decreases in mu and delta opioid receptors in the superficial dorsal horn of the rat spinal cord following a large unilateral dorsal rhizotomy. Brain Res 1992; 578: 115–27PubMedCrossRef Besse D, Lombard MC, Besson JM. Time-related decreases in mu and delta opioid receptors in the superficial dorsal horn of the rat spinal cord following a large unilateral dorsal rhizotomy. Brain Res 1992; 578: 115–27PubMedCrossRef
49.
go back to reference Hohmann AG, Herkenham M. Regulation of cannabinoid and mu opioid receptors in rat lumbar spinal cord following neonatal capsaicin treatment Neurosci Lett 1998; 252: 13–16 Hohmann AG, Herkenham M. Regulation of cannabinoid and mu opioid receptors in rat lumbar spinal cord following neonatal capsaicin treatment Neurosci Lett 1998; 252: 13–16
50.
go back to reference Farquhar-Smith WP, Egertova M, Bradbury EJ, et al. Cannabinoid CB(1) receptor expression in rat spinal cord. Mol Cell Neurosci 2000; 15: 510–21PubMedCrossRef Farquhar-Smith WP, Egertova M, Bradbury EJ, et al. Cannabinoid CB(1) receptor expression in rat spinal cord. Mol Cell Neurosci 2000; 15: 510–21PubMedCrossRef
51.
go back to reference Siegling A, Hofmann HA, Denzer D, et al. Cannabinoid CB(1) receptor upregulation in a rat model of chronic neuropathic pain. Eur Pharmacol 2001; 415: R5–7CrossRef Siegling A, Hofmann HA, Denzer D, et al. Cannabinoid CB(1) receptor upregulation in a rat model of chronic neuropathic pain. Eur Pharmacol 2001; 415: R5–7CrossRef
52.
go back to reference Monhemius R, Azami J, Green DL, et al. CB1 receptor mediated analgesia from the nucleus reticularis gigantocellularis pars alpha is activated in an animal model of neuropathic pain. Brain Res 2001; 908: 67–74PubMedCrossRef Monhemius R, Azami J, Green DL, et al. CB1 receptor mediated analgesia from the nucleus reticularis gigantocellularis pars alpha is activated in an animal model of neuropathic pain. Brain Res 2001; 908: 67–74PubMedCrossRef
53.
go back to reference Zhang J, Hoffert C, Vu HK, et al. Induction of CB2 receptor expression in the rat spinal cord of neuropathic but not inflammatory chronic pain models. Eur J Neurosci 2003; 17(12): 2750–4PubMedCrossRef Zhang J, Hoffert C, Vu HK, et al. Induction of CB2 receptor expression in the rat spinal cord of neuropathic but not inflammatory chronic pain models. Eur J Neurosci 2003; 17(12): 2750–4PubMedCrossRef
54.
go back to reference Bennett GJ, Xie YK. A peripheral mononeuropathy in rat that produces disorders of pain sensation like those seen in man. Pain 1988; 33(1): 87–107PubMedCrossRef Bennett GJ, Xie YK. A peripheral mononeuropathy in rat that produces disorders of pain sensation like those seen in man. Pain 1988; 33(1): 87–107PubMedCrossRef
55.
go back to reference Seltzer Z, Dubner R, Shir Y. A novel behavioral model of neuropathic pain disorders produced in rats by partial sciatic nerve injury. Pain 1990; 43(2): 205–18PubMedCrossRef Seltzer Z, Dubner R, Shir Y. A novel behavioral model of neuropathic pain disorders produced in rats by partial sciatic nerve injury. Pain 1990; 43(2): 205–18PubMedCrossRef
56.
go back to reference Burchiel KJ, Russell LC, Lee RP, et al. Spontaneous activity of primary afferent neurons in diabetic BB/Wistar rats: a possible mechanism of chronic diabetic neuropathic pain. Diabetes 1985; 34(11): 1210–3PubMedCrossRef Burchiel KJ, Russell LC, Lee RP, et al. Spontaneous activity of primary afferent neurons in diabetic BB/Wistar rats: a possible mechanism of chronic diabetic neuropathic pain. Diabetes 1985; 34(11): 1210–3PubMedCrossRef
57.
go back to reference Polomano RC, Mannes AJ, Clark US, et al. A painful peripheral neuropathy in the rat produced by the chemotherapeutic drug, paclitaxel. Pain 2001; 94(3): 293–304PubMedCrossRef Polomano RC, Mannes AJ, Clark US, et al. A painful peripheral neuropathy in the rat produced by the chemotherapeutic drug, paclitaxel. Pain 2001; 94(3): 293–304PubMedCrossRef
58.
go back to reference Tanner KD, Reichling DB, Levine JD. Nociceptor hyper-responsiveness during vincristine-induced painful peripheral neuropathy in the rat. J Neurosci 1998; 18(16): 6480–91PubMed Tanner KD, Reichling DB, Levine JD. Nociceptor hyper-responsiveness during vincristine-induced painful peripheral neuropathy in the rat. J Neurosci 1998; 18(16): 6480–91PubMed
59.
go back to reference Shinoda K, Hruby VJ, Porreca F. Antihyperalgesic effects of loperamide in a model of rat neuropathic pain are mediated by peripheral delta-opioid receptors. Neurosci Lett 2007; 411(2): 143–6PubMedCrossRef Shinoda K, Hruby VJ, Porreca F. Antihyperalgesic effects of loperamide in a model of rat neuropathic pain are mediated by peripheral delta-opioid receptors. Neurosci Lett 2007; 411(2): 143–6PubMedCrossRef
60.
go back to reference Choi Y, Yoon YW, Na HS, et al. Behavioral signs of ongoing pain and cold allodynia in a rat model of neuropathic pain. Pain 1994; 59(3): 369–76PubMedCrossRef Choi Y, Yoon YW, Na HS, et al. Behavioral signs of ongoing pain and cold allodynia in a rat model of neuropathic pain. Pain 1994; 59(3): 369–76PubMedCrossRef
61.
go back to reference Pascual D, Goicoechea C, Suardiaz M, et al. A cannabinoid agonist, WIN 55,212-2, reduces neuropathic nociception induced by paclitaxel in rats. Pain 2005; 118(1–2): 23–34PubMedCrossRef Pascual D, Goicoechea C, Suardiaz M, et al. A cannabinoid agonist, WIN 55,212-2, reduces neuropathic nociception induced by paclitaxel in rats. Pain 2005; 118(1–2): 23–34PubMedCrossRef
62.
go back to reference Dogrul A, Gul H, Yildiz O, et al. Cannabinoids blocks tactile allodynia in diabetic mice without attenuation of its antinociceptive effect. Neurosci Lett 2004; 368: 82–6PubMedCrossRef Dogrul A, Gul H, Yildiz O, et al. Cannabinoids blocks tactile allodynia in diabetic mice without attenuation of its antinociceptive effect. Neurosci Lett 2004; 368: 82–6PubMedCrossRef
63.
go back to reference Wood HC. Treatise on therapeutics. Philadelphia (PA): JB Lippincott and Co., 1886 Wood HC. Treatise on therapeutics. Philadelphia (PA): JB Lippincott and Co., 1886
64.
go back to reference Hare HA, Chrystie W. A system of practical therapeutics. Vol. 3. Philadelphia (PA): Lee Brothers, 1892 Hare HA, Chrystie W. A system of practical therapeutics. Vol. 3. Philadelphia (PA): Lee Brothers, 1892
65.
go back to reference Corey S. Recent developments in the therapeutic potential of cannabinoids. P R Health Sci J 2005; 24(1): 19–26PubMed Corey S. Recent developments in the therapeutic potential of cannabinoids. P R Health Sci J 2005; 24(1): 19–26PubMed
68.
go back to reference Campbell FA, Tramer MR, Carroll D, et al. Are cannabinoids an effective and safe treatment option in the management of pain? A qualitative systematic review. BMJ 2001; 323(7303): 13–6PubMedCrossRef Campbell FA, Tramer MR, Carroll D, et al. Are cannabinoids an effective and safe treatment option in the management of pain? A qualitative systematic review. BMJ 2001; 323(7303): 13–6PubMedCrossRef
69.
go back to reference Zajicek J, Fox P, Sanders H, et al. Cannabinoids for treatment of spasticity and other symptoms related to multiple sclerosis (CAMS study): multicentre randomised placebo-controlled trial. Lancet 2003; 362(9395): 1517–26PubMedCrossRef Zajicek J, Fox P, Sanders H, et al. Cannabinoids for treatment of spasticity and other symptoms related to multiple sclerosis (CAMS study): multicentre randomised placebo-controlled trial. Lancet 2003; 362(9395): 1517–26PubMedCrossRef
70.
go back to reference Wade DT, Robson P, House H, et al. A preliminary controlled study to determine whether whole-plant cannabis extracts can improve intractable neurogenic symptoms. Clinical Rehab 2003; 17(1): 21–9CrossRef Wade DT, Robson P, House H, et al. A preliminary controlled study to determine whether whole-plant cannabis extracts can improve intractable neurogenic symptoms. Clinical Rehab 2003; 17(1): 21–9CrossRef
71.
go back to reference Rog DJ, Nurmikko TJ, Friede T, et al. Randomized, controlled trial of cannabis-based medicine in central pain in multiple sclerosis. Neurology 2005; 65(6): 812–9PubMedCrossRef Rog DJ, Nurmikko TJ, Friede T, et al. Randomized, controlled trial of cannabis-based medicine in central pain in multiple sclerosis. Neurology 2005; 65(6): 812–9PubMedCrossRef
72.
go back to reference Svendsen KB, Jensen TS, Bach FW. Does the cannabinoid dronabinol reduce central pain in multiple sclerosis? Randomised, double-blind, placebo-controlled, crossover trial. BMJ 2004; 329(7460): 253–7PubMedCrossRef Svendsen KB, Jensen TS, Bach FW. Does the cannabinoid dronabinol reduce central pain in multiple sclerosis? Randomised, double-blind, placebo-controlled, crossover trial. BMJ 2004; 329(7460): 253–7PubMedCrossRef
73.
go back to reference Wissel J, Haydn T, Müller J, et al. Low dose treatment with the synthetic cannabinoid nabilone significantly reduces spasticity-related pain: a double-blind placebo-controlled cross-over trial. J Neurol 2006; 253(10): 1337–41PubMedCrossRef Wissel J, Haydn T, Müller J, et al. Low dose treatment with the synthetic cannabinoid nabilone significantly reduces spasticity-related pain: a double-blind placebo-controlled cross-over trial. J Neurol 2006; 253(10): 1337–41PubMedCrossRef
74.
go back to reference Berman JS, Symonds C, Birch R. Efficacy of two cannabis based medicinal extracts for relief of central neuropathic pain from brachial plexus avulsion: results of a randomised controlled trial. Pain 2004; 112(3): 299–36PubMedCrossRef Berman JS, Symonds C, Birch R. Efficacy of two cannabis based medicinal extracts for relief of central neuropathic pain from brachial plexus avulsion: results of a randomised controlled trial. Pain 2004; 112(3): 299–36PubMedCrossRef
75.
go back to reference Abrams DI, Jay CA, Shade SB, et al. Cannabis in painful HIV-associated sensory neuropathy: a randomized placebo-controlled trial. Neurology 2007; 68(7): 515–21PubMedCrossRef Abrams DI, Jay CA, Shade SB, et al. Cannabis in painful HIV-associated sensory neuropathy: a randomized placebo-controlled trial. Neurology 2007; 68(7): 515–21PubMedCrossRef
76.
go back to reference Karst M, Salim K, Burstein S, et al. Analgesic effect of the synthetic cannabinoid CT-3 on chronic neuropathic pain: a randomized controlled trial. JAMA 2003; 290(13): 1757–62PubMedCrossRef Karst M, Salim K, Burstein S, et al. Analgesic effect of the synthetic cannabinoid CT-3 on chronic neuropathic pain: a randomized controlled trial. JAMA 2003; 290(13): 1757–62PubMedCrossRef
77.
go back to reference Salim K, Schneider U, Burstein S, et al. Pain measurements and side effect profile of the novel cannabinoid ajulemic acid. Neuropharmacology 2005; 48(8): 1164–71PubMedCrossRef Salim K, Schneider U, Burstein S, et al. Pain measurements and side effect profile of the novel cannabinoid ajulemic acid. Neuropharmacology 2005; 48(8): 1164–71PubMedCrossRef
78.
go back to reference Pinsger M, Schimetta W, Volc D, et al. Benefits of an add-on treatment with the synthetic cannabinomimetic nabilone on patients with chronic pain: a randomized controlled trial. Wien Klin Worchenschr 2006; 118(11–12): 327–35CrossRef Pinsger M, Schimetta W, Volc D, et al. Benefits of an add-on treatment with the synthetic cannabinomimetic nabilone on patients with chronic pain: a randomized controlled trial. Wien Klin Worchenschr 2006; 118(11–12): 327–35CrossRef
79.
go back to reference Zajicek JP, Sanders HP, Wright DE, et al. Cannbinoids and multiple sclerosis (CAMS) study: safety and efficacy data for 12 month follow up. J Neurol Neurosurg Psychiatry 2005; 76: 1664–9PubMedCrossRef Zajicek JP, Sanders HP, Wright DE, et al. Cannbinoids and multiple sclerosis (CAMS) study: safety and efficacy data for 12 month follow up. J Neurol Neurosurg Psychiatry 2005; 76: 1664–9PubMedCrossRef
80.
go back to reference Petersen KL, Rowbotham MC. A new human experimental pain model: the heat/capsaicin sensitization model [published erratum appears in Neuroreport 2002 Jan 21; 13 (1): inside back cover]. Neuroreport 1999; 10(7): 1511–6PubMedCrossRef Petersen KL, Rowbotham MC. A new human experimental pain model: the heat/capsaicin sensitization model [published erratum appears in Neuroreport 2002 Jan 21; 13 (1): inside back cover]. Neuroreport 1999; 10(7): 1511–6PubMedCrossRef
81.
go back to reference Burstein S. Ajulemic acid (IP-751): synthesis, proof of principle, toxicity studies, and clinical trials. AAPS J 2005; 7(1): E143–8PubMedCrossRef Burstein S. Ajulemic acid (IP-751): synthesis, proof of principle, toxicity studies, and clinical trials. AAPS J 2005; 7(1): E143–8PubMedCrossRef
82.
go back to reference Vann RE, Cook CD, Martin BR, et al. Cannabimimetic properties of ajulemic acid. J Pharmacol Exp Ther 2007; 320(2): 678–86PubMedCrossRef Vann RE, Cook CD, Martin BR, et al. Cannabimimetic properties of ajulemic acid. J Pharmacol Exp Ther 2007; 320(2): 678–86PubMedCrossRef
83.
go back to reference Crippa JA, Zuardi AW, Garrido GE, et al. Effects of cannabidiol (CBD) on regional cerebral blood flow. Neuropsychopharmacology 2004; 29(2): 417–26PubMedCrossRef Crippa JA, Zuardi AW, Garrido GE, et al. Effects of cannabidiol (CBD) on regional cerebral blood flow. Neuropsychopharmacology 2004; 29(2): 417–26PubMedCrossRef
84.
go back to reference Wade DT, Makela P, Robson P, et al. Do cannabis-based medicinal extracts have general or specific effects on symptoms in multiple sclerosis? A double-blind, randomized, placebo-controlled study on 160 patients. Mult Scler 2004; 10(4): 434–41PubMedCrossRef Wade DT, Makela P, Robson P, et al. Do cannabis-based medicinal extracts have general or specific effects on symptoms in multiple sclerosis? A double-blind, randomized, placebo-controlled study on 160 patients. Mult Scler 2004; 10(4): 434–41PubMedCrossRef
85.
go back to reference Killestein J, Hoogervorst EL, Reif M, et al. Safety, tolerability, and efficacy of orally administered cannabinoids in MS. Neurology 2002; 58(9): 1404–7PubMedCrossRef Killestein J, Hoogervorst EL, Reif M, et al. Safety, tolerability, and efficacy of orally administered cannabinoids in MS. Neurology 2002; 58(9): 1404–7PubMedCrossRef
Metadata
Title
Role of Cannabinoids in the Management of Neuropathic Pain
Authors
Dr M. Isabel Martín Fontelles
Carlos Goicoechea García
Publication date
01-08-2008
Publisher
Springer International Publishing
Published in
CNS Drugs / Issue 8/2008
Print ISSN: 1172-7047
Electronic ISSN: 1179-1934
DOI
https://doi.org/10.2165/00023210-200822080-00003

Other articles of this Issue 8/2008

CNS Drugs 8/2008 Go to the issue