Skip to main content

Advertisement

Log in

The anti-inflammatory and analgesic effects of formulated full-spectrum cannabis extract in the treatment of neuropathic pain associated with multiple sclerosis

  • Review
  • Published:
Inflammation Research Aims and scope Submit manuscript

Abstract

Purpose

Cannabis has been used for thousands of years in many cultures for the treatment of several ailments including pain. The benefits of cannabis are mediated largely by cannabinoids, the most prominent of which are tetrahydrocannabinol (THC) and cannabidiol (CBD). As such, THC and/or CBD have been investigated in clinical studies for the treatment of many conditions including neuropathic pain and acute or chronic inflammation. While a plethora of studies have examined the biochemical effects of purified THC and/or CBD, only a few have focused on the effects of full-spectrum cannabis plant extract. Accordingly, studies using purified THC or CBD may not accurately reflect the potential health benefits of full-spectrum cannabis extracts. Indeed, the cannabis plant produces a wide range of cannabinoids, terpenes, flavonoids, and other bioactive molecules which are likely to contribute to the different biological effects. The presence of all these bioactive molecules in cannabis extracts has garnered much attention of late especially with regard to their potential role in the treatment of neuropathic pain associated with multiple sclerosis.

Methods

Literature review was performed to further understand the effect of clinically used full-spectrum cannabis extract in patients with multiple sclerosis.

Results

Herein, the current knowledge about the potential beneficial effects of existing products of full-spectrum cannabis extract in clinical studies involving patients with multiple sclerosis is extensively reviewed. In addition, the possible adverse effects associated with cannabis use is discussed along with how the method of extraction and the delivery mechanisms of different cannabis extracts contribute to the pharmacokinetic and biological effects of full-spectrum cannabis extracts.Herein, the current knowledge about the potential beneficial effects of existing products of full-spectrum cannabis extract in clinical studies involving patients with multiple sclerosis is extensively reviewed. In addition, the possible adverse effects associated with cannabis use is discussed along with how the method of extraction and the delivery mechanisms of different cannabis extracts contribute to the pharmacokinetic and biological effects of full-spectrum cannabis extracts.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

References

  1. Hagemeier K, Bruck W, Kuhlmann T. Multiple sclerosis—remyelination failure as a cause of disease progression. Histol Histopathol. 2012;27:277–87.

    CAS  PubMed  Google Scholar 

  2. Koch-Henriksen N, Sorensen PS. The changing demographic pattern of multiple sclerosis epidemiology. Lancet Neurol. 2010;9:520–32.

    PubMed  Google Scholar 

  3. Dhanani NM, Caruso TJ, Carinci AJ. Complementary and alternative medicine for pain: an evidence-based review. Curr Pain Headache Rep. 2011;15:39–46.

    PubMed  Google Scholar 

  4. Dimitrov LG, Turner B. What's new in multiple sclerosis? Br J Gen Pract. 2014;64:612–3.

    PubMed  PubMed Central  Google Scholar 

  5. Abel EL. Cannabis in the Ancient World. Marihuana: the first twelve thousand years 1980; New York City: Plenum Publishers.

  6. Clendinning J. Observations on the medicinal properties of the Cannabis sativa of India. Med-Chir Trans. 1843;26:188–21010.

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Ashton CH. Biomedical benefits of cannabinoids? Addict Biol. 1999;4:111–26.

    CAS  PubMed  Google Scholar 

  8. Burstein S. Cannabidiol (CBD) and its analogs: a review of their effects on inflammation. Bioorg Med Chem. 2015;23:1377–85.

    CAS  PubMed  Google Scholar 

  9. Holdcroft A, Maze M, Dore C, Tebbs S, Thompson S. A multicenter dose-escalation study of the analgesic and adverse effects of an oral cannabis extract (Cannador) for postoperative pain management. Anesthesiology. 2006;104:1040–6.

    CAS  PubMed  Google Scholar 

  10. Rog DJ, Nurmikko TJ, Friede T, Young CA. Randomized, controlled trial of cannabis-based medicine in central pain in multiple sclerosis. Neurology. 2005;65:812–9.

    PubMed  Google Scholar 

  11. 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;112:299–306.

    PubMed  Google Scholar 

  12. Lentela DE, Kamal BS, Kamal F. Cannabis and the anxiety of fragmentation—a systems approach for finding an anxiolytic Cannabis chemotype. Front Neurosci. 2018. https://doi.org/10.3389/fnins.2018.00730 (in press).

    Article  Google Scholar 

  13. Booth JK, Page JE, Bohlmann J. Terpene synthases from Cannabis sativa. PLoS ONE. 2017;12:e0173911.

    PubMed  PubMed Central  Google Scholar 

  14. Wagner H, Ulrich-Merzenich G. Synergy research: approaching a new generation of phytopharmaceuticals. Phytomedicine. 2009;16:97–110.

    CAS  PubMed  Google Scholar 

  15. Chen JW, Borgelt LM, Blackmer AB. Epidiolex (Cannabidiol): a new hope for patients with dravet or lennox–gastaut syndromes. Ann Pharmacother. 2019;53(6):603–11. https://doi.org/10.1177/1060028018822124.

    Article  CAS  PubMed  Google Scholar 

  16. Markova J, Essner U, Akmaz B, Marinelli M, Trompke C, Lentschat A, et al. Sativex((R)) as add-on therapy vs. further optimized first-line ANTispastics (SAVANT) in resistant multiple sclerosis spasticity: a double-blind, placebo-controlled randomised clinical trial. Int J Neurosci. 2019;129(2):119–28. https://doi.org/10.1080/00207454.2018.1481066.

    Article  CAS  PubMed  Google Scholar 

  17. Comelli F, Giagnoni G, Bettoni I, Colleoni M, Costa B. Antihyperalgesic effect of a Cannabis sativa extract in a rat model of neuropathic pain: mechanisms involved. Phytother Res. 2008;22:1017–24.

    PubMed  Google Scholar 

  18. May MB, Glode AE. Dronabinol for chemotherapy-induced nausea and vomiting unresponsive to antiemetics. Cancer Manag Res. 2016;8:49–55.

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Klumpers LE, Beumer TL, van Hasselt JG, Lipplaa A, Karger LB, Kleinloog HD, et al. Novel delta(9)-tetrahydrocannabinol formulation Namisol(R) has beneficial pharmacokinetics and promising pharmacodynamic effects. Br J Clin Pharmacol. 2012;74:42–53.

    CAS  PubMed  Google Scholar 

  20. van Amerongen G, Kanhai K, Baakman AC, Heuberger J, Klaassen E, Beumer TL, et al. Effects on spasticity and neuropathic pain of an oral formulation of delta 9-tetrahydrocannabinol in patients with progressive multiple sclerosis. Clin Ther. 2018;40:1467–82.

    PubMed  Google Scholar 

  21. Zajicek J, Ball S, Wright D, Vickery J, Nunn A, Miller D, et al. Effect of dronabinol on progression in progressive multiple sclerosis (CUPID): a randomised, placebo-controlled trial. Lancet Neurol. 2013;12:857–65.

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Schimrigk S, Marziniak M, Neubauer C, Kugler EM, Werner G, Abramov-Sommariva D. Dronabinol is a safe long-term treatment option for neuropathic pain patients. Eur Neurol. 2017;78:320–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Abuhasira R, Shbiro L, Landschaft Y. Medical use of cannabis and cannabinoids containing products—regulations in Europe and North America. Eur J Intern Med. 2018;49:2–6.

    PubMed  Google Scholar 

  24. Sastre-Garriga J, Vila C, Clissold S, Montalban X. THC and CBD oromucosal spray (Sativex(R)) in the management of spasticity associated with multiple sclerosis. Expert Rev Neurother. 2011;11:627–37.

    CAS  PubMed  Google Scholar 

  25. Perez J. Combined cannabinoid therapy via an oromucosal spray. Drugs Today. 2006;42:495–503.

    CAS  PubMed  Google Scholar 

  26. Langford RM, Mares J, Novotna A, Vachova M, Novakova I, Notcutt W, et al. A double-blind, randomized, placebo-controlled, parallel-group study of THC/CBD oromucosal spray in combination with the existing treatment regimen, in the relief of central neuropathic pain in patients with multiple sclerosis. J Neurol. 2013;260:984–97.

    CAS  PubMed  Google Scholar 

  27. Wade DT, Makela P, Robson P, House H, Bateman C. 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;10:434–41.

    CAS  PubMed  Google Scholar 

  28. Rog DJ, Nurmikko TJ, Young CA. Oromucosal delta9-tetrahydrocannabinol/cannabidiol for neuropathic pain associated with multiple sclerosis: an uncontrolled, open-label, 2-year extension trial. Clin Ther. 2007;29:2068–79.

    CAS  PubMed  Google Scholar 

  29. Novotna A, Mares J, Ratcliffe S, Novakova I, Vachova M, Zapletalova O, et al. A randomized, double-blind, placebo-controlled, parallel-group, enriched-design study of nabiximols* (Sativex((R)) ), as add-on therapy, in subjects with refractory spasticity caused by multiple sclerosis. Eur J Neurol. 2011;18:1122–31.

    CAS  PubMed  Google Scholar 

  30. Notcutt W, Langford R, Davies P, Ratcliffe S, Potts R. A placebo-controlled, parallel-group, randomized withdrawal study of subjects with symptoms of spasticity due to multiple sclerosis who are receiving long-term Sativex(R) (nabiximols). Mult Scler. 2012;18:219–28.

    CAS  PubMed  Google Scholar 

  31. Ferre L, Nuara A, Pavan G, Radaelli M, Moiola L, Rodegher M, et al. Efficacy and safety of nabiximols (Sativex((R))) on multiple sclerosis spasticity in a real-life Italian monocentric study. Neurol Sci. 2016;37:235–42.

    PubMed  Google Scholar 

  32. Flachenecker P, Henze T, Zettl UK. Nabiximols (THC/CBD oromucosal spray, Sativex(R)) in clinical practice–results of a multicenter, non-interventional study (MOVE 2) in patients with multiple sclerosis spasticity. Eur Neurol. 2014;71:271–9.

    CAS  PubMed  Google Scholar 

  33. Wade DT, Makela PM, House H, Bateman C, Robson P. Long-term use of a cannabis-based medicine in the treatment of spasticity and other symptoms in multiple sclerosis. Mult Scler. 2006;12:639–45.

    CAS  PubMed  Google Scholar 

  34. Barnes MP. Sativex: clinical efficacy and tolerability in the treatment of symptoms of multiple sclerosis and neuropathic pain. Expert Opin Pharmacother. 2006;7:607–15.

    CAS  PubMed  Google Scholar 

  35. Trojano M, Vila C. Effectiveness and tolerability of THC/CBD oromucosal spray for multiple sclerosis spasticity in italy: first data from a large observational study. Eur Neurol. 2015;74:178–85.

    CAS  PubMed  Google Scholar 

  36. Vermersch P, Trojano M. Tetrahydrocannabinol: cannabidiol oromucosal spray for multiple sclerosis-related resistant spasticity in daily practice. Eur Neurol. 2016;76:216–26.

    CAS  PubMed  Google Scholar 

  37. Collin C, Ehler E, Waberzinek G, Alsindi Z, Davies P, Powell K, et al. A double-blind, randomized, placebo-controlled, parallel-group study of Sativex, in subjects with symptoms of spasticity due to multiple sclerosis. Neurol Res. 2010;32:451–9.

    CAS  PubMed  Google Scholar 

  38. Collin C, Davies P, Mutiboko IK, Ratcliffe S, Sativex Spasticity in MSSG. Randomized controlled trial of cannabis-based medicine in spasticity caused by multiple sclerosis. Eur J Neurol. 2007;14:290–6.

    CAS  PubMed  Google Scholar 

  39. Russo M, Calabro RS, Naro A, Sessa E, Rifici C, D'Aleo G, et al. Sativex in the management of multiple sclerosis-related spasticity: role of the corticospinal modulation. Neural Plast. 2015;2015:656582.

    PubMed  PubMed Central  Google Scholar 

  40. Squintani G, Donato F, Turri M, Deotto L, Teatini F, Moretto G, et al. Cortical and spinal excitability in patients with multiple sclerosis and spasticity after oromucosal cannabinoid spray. J Neurol Sci. 2016;370:263–8.

    CAS  PubMed  Google Scholar 

  41. Russo M, Naro A, Leo A, Sessa E, D'Aleo G, Bramanti P, et al. Evaluating Sativex(R) in neuropathic pain management: a clinical and neurophysiological assessment in multiple sclerosis. Pain Med. 2016;17:1145–54.

    PubMed  Google Scholar 

  42. Sorosina M, Clarelli F, Ferre L, Osiceanu AM, Unal NT, Mascia E, et al. Clinical response to Nabiximols correlates with the downregulation of immune pathways in multiple sclerosis. Eur J Neurol. 2018;25:934–e70.

    CAS  PubMed  Google Scholar 

  43. Turri M, Teatini F, Donato F, Zanette G, Tugnoli V, Deotto L, et al. Pain modulation after oromucosal cannabinoid spray (SATIVEX((R))) in patients with multiple sclerosis: a study with quantitative sensory testing and laser-evoked potentials. Medicines (Basel). 2018;5(3):pii: E59. https://doi.org/10.3390/medicines5030059.

    Article  CAS  Google Scholar 

  44. Lemberger L, Silberstein SD, Axelrod J, Kopin IJ. Marihuana: studies on the disposition and metabolism of delta-9-tetrahydrocannabinol in man. Science. 1970;170:1320–2.

    CAS  PubMed  Google Scholar 

  45. Schlicker E, Kathmann M. Modulation of transmitter release via presynaptic cannabinoid receptors. Trends Pharmacol Sci. 2001;22:565–72.

    CAS  PubMed  Google Scholar 

  46. Neurobiology BM. How cannabinoids work in the brain. Science. 2001;291:2530–1.

    Google Scholar 

  47. Comelli F, Bettoni I, Colleoni M, Giagnoni G, Costa B. Beneficial effects of a Cannabis sativa extract treatment on diabetes-induced neuropathy and oxidative stress. Phytother Res. 2009;23:1678–84.

    PubMed  Google Scholar 

  48. Ibrahim MM, Deng H, Zvonok A, Cockayne DA, Kwan J, Mata HP, et al. Activation of CB2 cannabinoid receptors by AM1241 inhibits experimental neuropathic pain: pain inhibition by receptors not present in the CNS. Proc Natl Acad Sci USA. 2003;100:10529–33.

    CAS  PubMed  Google Scholar 

  49. Ross HR, Napier I, Connor M. Inhibition of recombinant human T-type calcium channels by delta 9-tetrahydrocannabinol and cannabidiol. J Biol Chem. 2008;283:16124–34.

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Aragona M, Onesti E, Tomassini V, Conte A, Gupta S, Gilio F, et al. Psychopathological and cognitive effects of therapeutic cannabinoids in multiple sclerosis: a double-blind, placebo-controlled, crossover study. Clin Neuropharmacol. 2009;32:41–7.

    CAS  PubMed  Google Scholar 

  51. Massi P, Valenti M, Vaccani A, Gasperi V, Perletti G, Marras E, et al. 5-Lipoxygenase and anandamide hydrolase (FAAH) mediate the antitumor activity of cannabidiol, a non-psychoactive cannabinoid. J Neurochem. 2008;104:1091–100.

    CAS  PubMed  Google Scholar 

  52. Karschner EL, Darwin WD, Goodwin RS, Wright S, Huestis MA. Plasma cannabinoid pharmacokinetics following controlled oral delta 9-tetrahydrocannabinol and oromucosal cannabis extract administration. Clin Chem. 2011;57:66–75.

    CAS  PubMed  Google Scholar 

  53. Zajicek J, Fox P, Sanders H, Wright D, Vickery J, Nunn A, et al. Cannabinoids for treatment of spasticity and other symptoms related to multiple sclerosis (CAMS study): multicentre randomised placebo-controlled trial. Lancet. 2003;362:1517–26.

    CAS  PubMed  Google Scholar 

  54. Zajicek JP, Sanders HP, Wright DE, Vickery PJ, Ingram WM, Reilly SM, et al. Cannabinoids in multiple sclerosis (CAMS) study: safety and efficacy data for 12 months follow up. J Neurol Neurosurg Psychiatry. 2005;76:1664–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Zajicek JP, Hobart JC, Slade A, Barnes D, Mattison PG, Group MR. Multiple sclerosis and extract of cannabis: results of the MUSEC trial. J Neurol Neurosurg Psychiatry. 2012;83:1125–32.

    PubMed  Google Scholar 

  56. Urits I, Borchart M, Hasegawa M, Kochanski J, Orhurhu V, Viswanath O. An update of current cannabis-based pharmaceuticals in pain medicine. Pain Therapy. 2019;8:41–51.

    PubMed  Google Scholar 

  57. Babalonis S, Haney M, Malcolm RJ, Lofwall MR, Votaw VR, Sparenborg S, et al. Oral cannabidiol does not produce a signal for abuse liability in frequent marijuana smokers. Drug Alcohol Depend. 2017;172:9–13.

    CAS  PubMed  Google Scholar 

  58. Thiele E, Marsh E, Mazurkiewicz-Beldzinska M, Halford JJ, Gunning B, Devinsky O, et al. Cannabidiol in patients with Lennox-Gastaut syndrome: interim analysis of an open-label extension study. Epilepsia. 2019;60(3):419–28. https://doi.org/10.1111/epi.14670.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Serpell MG, Notcutt W, Collin C. Sativex long-term use: an open-label trial in patients with spasticity due to multiple sclerosis. J Neurol. 2013;260:285–95.

    PubMed  Google Scholar 

  60. Patti F, Messina S, Solaro C, Amato MP, Bergamaschi R, Bonavita S, et al. Efficacy and safety of cannabinoid oromucosal spray for multiple sclerosis spasticity. J Neurol Neurosurg Psychiatry. 2016;87:944–51.

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Haupts M, Vila C, Jonas A, Witte K, Alvarez-Ossorio L. Influence of previous failed antispasticity therapy on the efficacy and tolerability of THC:CBD oromucosal spray for multiple sclerosis spasticity. Eur Neurol. 2016;75:236–43.

    CAS  PubMed  Google Scholar 

  62. Leocani L, Nuara A, Houdayer E, Schiavetti I, Del Carro U, Amadio S, et al. Sativex((R)) and clinical-neurophysiological measures of spasticity in progressive multiple sclerosis. J Neurol. 2015;262:2520–7.

    CAS  PubMed  Google Scholar 

  63. Constantinescu CS, Gershkovich P. Therapeutic cannabinoids in multiple sclerosis: immunomodulation revisited. Eur J Neurol. 2018;25:905–6.

    CAS  PubMed  Google Scholar 

  64. Etges T, Karolia K, Grint T, Taylor A, Lauder H, Daka B, et al. An observational postmarketing safety registry of patients in the UK, Germany, and Switzerland who have been prescribed Sativex((R)) (THC:CBD, nabiximols) oromucosal spray. Ther Clin Risk Manag. 2016;12:1667–755.

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Freeman RM, Adekanmi O, Waterfield MR, Waterfield AE, Wright D, Zajicek J. The effect of cannabis on urge incontinence in patients with multiple sclerosis: a multicentre, randomised placebo-controlled trial (CAMS-LUTS). Int Urogynecol J Pelvic Floor Dysfunct. 2006;17:636–41.

    CAS  PubMed  Google Scholar 

  66. Vaney C, Heinzel-Gutenbrunner M, Jobin P, Tschopp F, Gattlen B, Hagen U, et al. Efficacy, safety and tolerability of an orally administered cannabis extract in the treatment of spasticity in patients with multiple sclerosis: a randomized, double-blind, placebo-controlled, crossover study. Mult Scler. 2004;10:417–24.

    CAS  PubMed  Google Scholar 

  67. Buggy DJ, Toogood L, Maric S, Sharpe P, Lambert DG, Rowbotham DJ. Lack of analgesic efficacy of oral delta-9-tetrahydrocannabinol in postoperative pain. Pain. 2003;106:169–72.

    CAS  PubMed  Google Scholar 

  68. Naef M, Curatolo M, Petersen-Felix S, Arendt-Nielsen L, Zbinden A, Brenneisen R. The analgesic effect of oral delta-9-tetrahydrocannabinol (THC), morphine, and a THC–morphine combination in healthy subjects under experimental pain conditions. Pain. 2003;105:79–88.

    CAS  PubMed  Google Scholar 

  69. Russo EB. Taming THC: potential cannabis synergy and phytocannabinoid—terpenoid entourage effects. Br J Pharmacol. 2011;163:1344–64.

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Mechoulam R, Parker LA. The endocannabinoid system and the brain. Annu Rev Psychol. 2013;64:21–47.

    PubMed  Google Scholar 

  71. Baron EP. Medicinal properties of cannabinoids, terpenes, and flavonoids in cannabis, and benefits in migraine, headache, and pain: an update on current evidence and cannabis science. Headache. 2018;58:1139–86.

    PubMed  Google Scholar 

  72. Zgair A, Lee JB, Wong JCM, Taha DA, Aram J, Di Virgilio D, et al. Oral administration of cannabis with lipids leads to high levels of cannabinoids in the intestinal lymphatic system and prominent immunomodulation. Sci Rep. 2017;7:14542.

    PubMed  PubMed Central  Google Scholar 

  73. Huestis MA. Human cannabinoid pharmacokinetics. Chem Biodivers. 2007;4:1770–804.

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Turner CE, Elsohly MA, Boeren EG. Constituents of Cannabis sativa L. XVII. A review of the natural constituents. J Nat Prod. 1980;43:169–234.

    CAS  PubMed  Google Scholar 

  75. Tanasescu R, Constantinescu CS. Pharmacokinetic evaluation of nabiximols for the treatment of multiple sclerosis pain. Expert Opin Drug Metab Toxicol. 2013;9:1219–28.

    CAS  PubMed  Google Scholar 

  76. Rousseaux C, Thuru X, Gelot A, Barnich N, Neut C, Dubuquoy L, et al. Lactobacillus acidophilus modulates intestinal pain and induces opioid and cannabinoid receptors. Nat Med. 2007;13:35–7.

    CAS  PubMed  Google Scholar 

  77. Cluny NL, Keenan CM, Reimer RA, Le Foll B, Sharkey KA. Prevention of diet-induced obesity effects on body weight and gut microbiota in mice treated chronically with delta 9-tetrahydrocannabinol. PLoS ONE. 2015;10:e0144270.

    PubMed  PubMed Central  Google Scholar 

  78. Ruth MR, Field CJ. The immune modifying effects of amino acids on gut-associated lymphoid tissue. J Anim Sci Biotechnol. 2013;4:27.

    PubMed  PubMed Central  Google Scholar 

  79. Yiangou Y, Facer P, Durrenberger P, Chessell IP, Naylor A, Bountra C, et al. COX-2, CB2 and P2X7-immunoreactivities are increased in activated microglial cells/macrophages of multiple sclerosis and amyotrophic lateral sclerosis spinal cord. BMC Neurol. 2006;6:12.

    PubMed  PubMed Central  Google Scholar 

  80. Zgair A, Wong JC, Lee JB, Mistry J, Sivak O, Wasan KM, et al. Dietary fats and pharmaceutical lipid excipients increase systemic exposure to orally administered cannabis and cannabis-based medicines. Am J Transl Res. 2016;8:3448–59.

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Institute of Medicine (US), Joy JE, Watson SJ Jr, Benson JA Jr, editors. Marijuana and medicine: assessing the science base. Washington, DC: National Academies Press; 1999.

    Google Scholar 

  82. Abrams DI, Jay CA, Shade SB, Vizoso H, Reda H, Press S, et al. Cannabis in painful HIV-associated sensory neuropathy: a randomized placebo-controlled trial. Neurology. 2007;68:515–21.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by a grant from Mitacs (Grant No. IT10775) Accelerate to JRBD. JRBD is a Canada Research Chair in Molecular Medicine. ZHM is the recipient of Mitacs Accelerate fellowship and the Alberta Innovates Health Solution post-doctoral fellowship award.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Jason R. B. Dyck.

Ethics declarations

Conflict of interest

The authors have declared that no conflict of interest exists with this review.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Maayah, Z.H., Takahara, S., Ferdaoussi, M. et al. The anti-inflammatory and analgesic effects of formulated full-spectrum cannabis extract in the treatment of neuropathic pain associated with multiple sclerosis. Inflamm. Res. 69, 549–558 (2020). https://doi.org/10.1007/s00011-020-01341-1

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00011-020-01341-1

Keywords

Navigation