Skip to main content
Log in

Cannabinoids: Current and Future Options to Treat Chronic and Chemotherapy-Induced Neuropathic Pain

  • Review Article
  • Published:
Drugs Aims and scope Submit manuscript

Abstract

Increases in cancer diagnosis have tremendous negative impacts on patients and their families, and major societal and economic costs. The beneficial effect of chemotherapeutic agents on tumor suppression comes with major unwanted side effects such as weight and hair loss, nausea and vomiting, and neuropathic pain. Chemotherapy-induced peripheral neuropathy (CIPN), which can include both painful and non-painful symptoms, can persist 6 months or longer after the patient’s last chemotherapeutic treatment. These peripheral sensory and motor deficits are poorly treated by our current analgesics with limited effectiveness. Therefore, the development of novel treatment strategies is an important preclinical research focus and an urgent need for patients. Approaches to prevent CIPN have yielded disappointing results since these compounds may interfere with the anti-tumor properties of chemotherapeutic agents. Nevertheless, the first (serotonin noradrenaline reuptake inhibitors [SNRIs], anticonvulsants, tricyclic antidepressants) and second (5% lidocaine patches, 8% capsaicin patches and weak opioids such as tramadol) lines of treatment for CIPN have shown some efficacy. The clinical challenge of CIPN management in cancer patients and the need to target novel therapies with long-term efficacy in alleviating CIPN are an ongoing focus of research. The endogenous cannabinoid system has shown great promise and efficacy in alleviating CIPN in preclinical and clinical studies. In this review, we will discuss the mechanisms through which the platinum, taxane, and vinca alkaloid classes of chemotherapeutics may produce CIPN and the potential therapeutic effect of drugs targeting the endocannabinoid system in preclinical and clinical studies, in addition to cannabinoid compounds diffuse mechanisms of action in alleviation of CIPN.

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
Fig. 2
Fig. 3
Fig. 4
Fig. 5

Similar content being viewed by others

References

  1. American Cancer Society. Cancer facts and figures 2018. Atlanta: American Cancer Society; 2018.

    Google Scholar 

  2. Grisold W, Cavaletti G, Windeback AJ. Peripheral neuropathies from chemotherapeutics and targets agents: diagnosis, treatment, and prevention. Neuro Oncol. 2012;14:45–54.

    Article  CAS  Google Scholar 

  3. Staff NP, Grisold A, Grisold W, Windebank AJ. Chemotherapy-induced peripheral neuropathy: a current review. Ann Neurol. 2017;81:772–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Windebank AJ, Grisold W. Chemotherapy-induced neuropathy. J Peripher Nerv Syst. 2008;13:27–46.

    Article  CAS  PubMed  Google Scholar 

  5. Pike CT, Birnbaum HG, Muehlenbein CE, Pohl GM, Natale RB. Healthcare costs and workloss burden of patients with chemotherapy-associated peripheral neuropathy in breast, ovarian, head and neck, and nonsmall cell lung cancer. Chemother Res Pract. 2012;2012:913848.

    PubMed  PubMed Central  Google Scholar 

  6. Seretny M, Currie GL, Sena ES, Ramnarine S, Grant R, MacLeod MR, et al. Incidence, prevalence, and predictors of chemotherapy-induced peripheral neuropathy: a systematic review and meta-analysis. Pain. 2014;155:2461–70.

    Article  PubMed  Google Scholar 

  7. Gaskin DJ, Richar P. The economic costs of pain in the United States. J Pain. 2012;13:715–24.

    Article  PubMed  Google Scholar 

  8. Nahin RL. Estimates of pain prevalence and severity in adults: United States, 2012. J Pain. 2015;2015(16):769–80.

    Article  Google Scholar 

  9. Fallon MT. Neuropathic pain in cancer. Br J Anaesth. 2013;111:105–11.

    Article  CAS  PubMed  Google Scholar 

  10. Kannarkat G, Lasher EE, Schiff D. Neurologic complications of chemotherapy agents. Curr Opin Neurol. 2007;20:19–25.

    Article  Google Scholar 

  11. Paice JA. Chronic treatment-related pain in cancer survivors. Pain. 2011;152:S84–9.

    Article  PubMed  Google Scholar 

  12. Flatters SJL, Dougherty PM, Colvin LA. Clinical and preclinical perspectives on chemotherapy-induced peripheral neuropathy (CIPN): a narrative review. Br J Anaesth. 2017;119:737–49.

    Article  CAS  PubMed  Google Scholar 

  13. Kim PY, Johnson CE. Chemotherapy-induced peripheral neuropathy: a review of recent findings. Curr Opin Anaesthesiol. 2017;30:570–6.

    Article  CAS  PubMed  Google Scholar 

  14. Wolf S, Barton D, Kottschade L, Grothey A, Loprinzi C. Chemotherapy-induced peripheral neuropathy: prevention and treatment strategies. Eur J Cancer. 2008;44:1507–15.

    Article  CAS  PubMed  Google Scholar 

  15. Cooper TE, Chen J, Wiffen PJ, Derry S, Carr DB, Aldington D, Cole P, Moore RA. Morphine for chronic neuropathic pain in adults. Cochrane Database Syst Rev. 2017. https://doi.org/10.1002/14651858.cd011669.pub2.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Fradkin M, Batash R, Elamleh S, Debi R, Schaffer P, Schaffer M, et al. Management of peripheral neuropathy induced by chemotherapy. Curr Med Chem. 2019. https://doi.org/10.2174/0929867326666190107163756.

    Article  PubMed  Google Scholar 

  17. Gilron I, Bailey JM, Tu D, Holden RR, Weaver DF, Houlden RL. Morphine, gabapentin, or their combination for neuropathic pain. N Engl J Med. 2005;352:1324–34.

    Article  CAS  PubMed  Google Scholar 

  18. Guindon J, Hohmann AG. The endocannabinoid system and pain. CNS Neurol Disord Drug Targets. 2009;8:403–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Rock EM, Parker LA. Cannabinoids as potential treatment for chemotherapy-induced nausea and vomiting. Front Pharmacol. 2016;7:221.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Whiting D. Synthetic cannabinoid receptor agonists: a heterogeneous class of novel psychoactive substance with emerging risk of psychosis. Evid Based Ment Health. 2015;18:110.

    Article  PubMed  Google Scholar 

  21. Balneaves LG, Alraja A, Ziemianski D, McCuaig F, Ware M. A national needs assessment of Canadian nurse practitioners regarding cannabis for therapeutic purposes. Cannabis Cannabinoid Res. 2018;3:66–73.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Ware MA, Wang T, Shapiro S, Collet JP. Cannabis for the Management of Pain: Assessment of Safety Study (COMPASS). J Pain. 2015;16:1233–42.

    Article  PubMed  Google Scholar 

  23. Hazekamp A, Ware MA, Muller-Vahl KR, Abrams D, Grotenhermen F. The medicinal use of cannabis and cannabinoids–an international cross-sectional survey on administration forms. J Psychoact Drugs. 2013;45:199–210.

    Article  Google Scholar 

  24. Lynch ME, Ware MA. Cannabinoids for the treatment of chronic non-cancer pain: an updated systematic review of randomized controlled trials. J Neuroimmune Pharmacol. 2015;10:293–301.

    Article  CAS  PubMed  Google Scholar 

  25. The National Academies of Sciences, Engineering and Medicine. The health effects of cannabis and cannabinoids. The current state of evidence and recommendations for research. 2017. http://www.nationalacademies.org. https://doi.org/10.17226/24625. https://www.ncbi.nlm.nih.gov/books/NBK423845/.

  26. Wishart DS, Feunang YD, Guo AC, Lo EJ, Marcu A, Grant JR, Sajed T, et al. DrugBank 5.0: a major update to the DrugBank database for 2018. Nucleic Acids Res. 2018;46:D1074–82.

    Article  CAS  PubMed  Google Scholar 

  27. Stover DG, Winer EP. Tailoring adjuvant chemotherapy regimens for patients with triple negative breast cancer. Breast. 2015;24:S132–5.

    Article  PubMed  Google Scholar 

  28. Yao H, He G, Yan S, Chen C, Song L, Rosol TJ, et al. Triple-negative breast cancer: is there a treatment on the horizon? Oncotarget. 2017;8:1913–24.

    PubMed  Google Scholar 

  29. Dasari S, Tchounwou PB. Cisplatin in cancer therapy: molecular mechanisms of action. Eur J Pharmacol. 2014;740:364–78.

    Article  CAS  PubMed  Google Scholar 

  30. Jordan MA, Wilson L. Microtubules as a target for anticancer drugs. Nat Rev Cancer. 2004;4:253–65.

    Article  CAS  PubMed  Google Scholar 

  31. Starobova H, Vetter I. Pathophysiology of chemotherapy-induced peripheral neuropathy. Front Mol Neurosci. 2017;10:174.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Boyette-Davis J, Dougherty PM. Protection against oxaliplatin-induced mechanical hyperalgesia and intraepidermal nerve fiber loss by minocycline. Exp Neurol. 2011;229:353–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Siau C, Xiao W, Bennett GJ. Paclitaxel- and vincristine-evoked painful peripheral neuropathies: loss of epidermal innervation and activation of Langerhans cells. Exp Neurol. 2006;201(2):507–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Di Cesare Mannelli L, Pacini A, Micheli L, Tani A, Zanardelli M, Ghelardini C. Glial role in oxaliplatin-induced neuropathic pain. Exp Neurol. 2014;261:22–33.

    Article  CAS  Google Scholar 

  35. Burgos E, Gomez-Nicola D, Pascual D, Martin MI, Nieto-Sampedro M, Goicoechea C. 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;682:62–72.

    Article  CAS  Google Scholar 

  36. Zhang L, Dermawan K, Jin M, Liu R, Zheng H, Xu L, et al. Differential impairment of regulatory T cells rather than effector T cells by paclitaxel-based chemotherapy. Clin Immunol. 2008;129:219–29.

    Article  CAS  PubMed  Google Scholar 

  37. Krukowski K, Eijkelkamp N, Laumet G, Hack CE, Li Y, Dougherty PM, et al. CD8 + T cells and endogenous IL-10 are required for resolution of chemotherapy-induced neuropathic pain. J Neurosci. 2016;36:11074–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Ledeboer A, Jekich BM, Sloane EM, Mahoney JH, Langer SJ, Milligan ED, et al. Intrathecal interleukin-10 gene therapy attenuates paclitaxel-induced mechanical allodynia and proinflammatory cytokine expression in dorsal root ganglia in rats. Brain Behav Immun. 2007;21:686–98.

    Article  CAS  PubMed  Google Scholar 

  39. Xu Z, Lv XA, Dai Q, Ge YQ, Xu J. Acute upregulation of neuronal mitochondrial type-1 cannabinoid receptor and it’s role in metabolic defects and neuronal apoptosis after TBI. Mol Brain. 2016;9:75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Li D, Huang Z-Z, Ling Y-Z, Wei J-Y, Cui Y, Zhang X-Z, et al. Up-regulation of CX3CL1 via nuclear factor-κB—dependent histone acetylation is involved in paclitaxel-induced peripheral neuropathy. Anesthesiology. 2015;122:1142–51.

    Article  CAS  PubMed  Google Scholar 

  41. Pevida M, Lastra A, Hidalgo A, Baamonde A, Menendez L. Spinal CCL2 and microglial activation are involved in paclitaxel-evoked cold hyperalgesia. Brain Res Bull. 2013;95:21–7.

    Article  CAS  PubMed  Google Scholar 

  42. Liu XJ, Zhang Y, Liu T, Xu ZZ, Park CK, Berta T, et al. Nociceptive neurons regulate innate and adaptive immunity and neuropathic pain through MyD88 adapter. Cell Res. 2014;24:1374–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Zhang H, Dougherty PM. Enhanced excitability of primary sensory neurons and altered gene expression of neuronal ion channels in dorsal root ganglion in paclitaxel-induced peripheral neuropathy. Anesthesiology. 2014;120:1463–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Gauchan P, Andoh T, Kato A, Kuraishi Y. Involvement of increased expression of transient receptor potential melastatin 8 in oxaliplatin-induced cold allodynia in mice. Neurosci Lett. 2009;458:93–5.

    Article  CAS  PubMed  Google Scholar 

  45. Descoeur J, Pereira V, Pizzoccaro A, Francois A, Ling B, Maffre V, et al. Oxaliplatin-induced cold hypersensitivity is due to remodelling of ion channel expression in nociceptors. EMBO Mol Med. 2011;3:266–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Nassini R, Gees M, Harrison S, De Siena G, Materazzi S, Moretto N, et al. Oxaliplatin elicits mechanical and cold allodynia in rodents via TRPA1 receptor stimulation. Pain. 2011;152:1621–31.

    Article  CAS  PubMed  Google Scholar 

  47. Ta LE, Bieber AJ, Carlton SM, Loprinzi CL, Low PA, Windebank AJ. Transient receptor potential vanilloid 1 is essential for cisplatin-induced heat hyperalgesia in mice. Mol Pain. 2010;6:15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Griffiths LA, Flatters SJ. Pharmacological modulation of the mitochondrial electron transport chain in paclitaxel-induced painful peripheral neuropathy. J Pain. 2015;16:981–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Jiang Y, Guo C, Vasko MR, Kelley MR. Implications of apurinic/apyrimidinic endonuclease in reactive oxygen signaling response after cisplatin treatment of dorsal root ganglion neurons. Cancer Res. 2008;68:6425–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Canta A, Pozzi E, Carozzi VA. Mitochondrial dysfunction in chemotherapy-induced peripheral neuropathy (CIPN). Toxics. 2015;3:198–223.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Guindon J, Walczak JS, Beaulieu P. Recent advances in the pharmacological management of pain. Drugs. 2007;67:2121–33.

    Article  CAS  PubMed  Google Scholar 

  52. Vardeh D, Mannion RJ, Woolf CJ. Toward a mechanism-based approach to pain diagnosis. J Pain. 2016;17:T50–69.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Karst M, Salim K, Burstein S, Conrad I, Hoy L, Schneider U. Analgesic effect of the synthetic cannabinoid CT-3 on chronic neuropathic pain: a randomized controlled trial. JAMA. 2003;290:1757–62.

    Article  CAS  PubMed  Google Scholar 

  54. Di Marzo V. Endocannabinoids and other fatty acid derivatives with cannabimimetic properties: biochemistry and possible physiopathological relevance. Biochim Biophys Acta. 1998;1392:153–75.

    Article  PubMed  Google Scholar 

  55. Piomelli D. The endocannabinoid system: a drug discovery perspective. Curr Opin Investig Drugs. 2005;6:672–9.

    CAS  PubMed  Google Scholar 

  56. Howlett AC, Breivogel CS, Childers SR, Deadwyler SA, Hampson RE, Porrino LJ. Cannabinoid physiology and pharmacology: 30 years of progress. Neuropharmacology. 2004;47:345–58.

    Article  CAS  PubMed  Google Scholar 

  57. Devane WA, Dysarz FA, Johnson MR, Melvin LS, Howlett AC. Determination and characterization of a cannabinoid receptor in rat brain. Mol Pharmacol. 1988;34:605–13.

    CAS  PubMed  Google Scholar 

  58. Matsuda LA, Lolait SJ, Brownstein MJ, Young AC, Bonner TI. Structure of a cannabinoid receptor and functional expression of the cloned cDNA. Nature. 1990;346:561–4.

    Article  CAS  PubMed  Google Scholar 

  59. Munro S, Thomas KL, Abu-Shaar M. Molecular characterization of a peripheral receptor for cannabinoids. Nature. 1993;365:61–5.

    Article  CAS  PubMed  Google Scholar 

  60. Pertwee RG. Cannabinoid receptors and pain. Prog Neurobiol. 2001;63:569–611.

    Article  CAS  PubMed  Google Scholar 

  61. Smart D, Gunthorpe MJ, Jerman JC, Nasir S, Gray J, Muir AI, et al. The endogenous lipid anandamide is a full agonist at the human vanilloid receptor (hVR1). Br J Pharmacol. 2000;129:227–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Mackie K, Hille B. Cannabinoids inhibit N-type calcium channels in neuroblastoma-glioma cells. Proc Natl Acad Sci USA. 1992;89:3825–9.

    Article  CAS  PubMed  Google Scholar 

  63. Howlett AC. The cannabinoid receptors. Prostaglandins Other Lipid Mediat. 2002;68–69:619–31.

    Article  PubMed  Google Scholar 

  64. Mackie K, Lai Y, Westenbroek R. Cannabinoids activate an inwardly rectifying potassium conductance and inhibit Q-type calcium currents in AtT20 cells transfected with rat brain cannabinoid receptor. J Neurosci. 1995;15:6552–61.

    Article  CAS  PubMed  Google Scholar 

  65. Pertwee RG. Pharmacological actions of cannabinoids. Handb Exp Pharmacol. 2005;168:1–51.

    Article  CAS  Google Scholar 

  66. Bridges D, Rice ASC, Egertová M, Elphick MR, Winter J, Michael GJ. Localisation of cannabinoid receptor 1 in rat dorsal root ganglion using in situ hybridisation and immunohistochemistry. Neuroscience. 2003;119:803–12.

    Article  CAS  PubMed  Google Scholar 

  67. Morisset V, Urban L. Cannabinoid-induced presynaptic inhibition of glutamatergic EPSCs in substantia gelatinosa neurons of the rat spinal cord. J Neurophysiol. 2001;86:40–8.

    Article  CAS  PubMed  Google Scholar 

  68. Martin WJ, Hohmann AG, Walker JM. Suppression of noxious stimulus-evoked activity in the ventral posterolateral nucleus of the thalamus by a cannabinoid agonist: correlation between electrophysiological and antinociceptive effects. J Neurosci. 1996;16:6601–11.

    Article  CAS  PubMed  Google Scholar 

  69. Martin WJ, Tsou K, Walker JM. Cannabinoid receptor-mediated inhibition of the rat tail-flick reflex after microinjection into the rostral ventromedial medulla. Neurosci Lett. 1998;242:33–6.

    Article  CAS  PubMed  Google Scholar 

  70. Herkenham M, Lynn AB, Johnson MR, Melvin LS, de Costa BR, Rice KC. Characterization and localization of cannabinoid receptors in rat brain: a quantitative in vitro autoradiographic study. J Neurosci. 1991;11:563–83.

    Article  CAS  PubMed  Google Scholar 

  71. Manning BH, Martin WJ, Meng ID. The rodent amygdala contributes to the production of cannabinoid-induced antinociception. Neuroscience. 2003;120:1157–70.

    Article  CAS  PubMed  Google Scholar 

  72. Lichtman AH, Cook SA, Martin BR. Investigation of brain sites mediating cannabinoid- induced antinociception in rats: evidence supporting periaqueductal gray involvement. J Pharmacol Exp Ther. 1996;276:585–93.

    CAS  PubMed  Google Scholar 

  73. Vera G, Cabezos PA, Martin MI, Abalo R. Characterization of cannabinoid-induced relief of neuropathic pain in a rat model of cisplatin-induced neuropathy. Pharmacol Biochem Behav. 2013;105:205–12.

    Article  CAS  PubMed  Google Scholar 

  74. Salaga M, Binienda A, Tichkule RB, Thakur GA, Makriyannis A, Storr M, et al. The novel peripherally active cannabinoid type 1 and serotonin type 3 receptor agonist AM9405 inhibits gastrointestinal motility and reduces abdominal pain in mouse models mimicking irritable bowel syndrome. Eur J Pharmacol. 2018;836:34–43.

    Article  CAS  PubMed  Google Scholar 

  75. Atwood B, Mackie K. CB2: a cannabinoid receptor with an identity crisis. Br J Pharmacol. 2010;160:467–79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Guindon J, Hohmann AG. Cannabinoid CB2 receptors: a therapeutic target for the treatment of inflammatory and neuropathic pain. Br J Pharmacol. 2008;153:319–34.

    Article  CAS  PubMed  Google Scholar 

  77. Deng L, Guindon J, Cornett BL, Makriyannis A, Mackie K, Hohmann AG. Chronic cannabinoid receptor 2 activation reverses paclitaxel neuropathy without tolerance or cannabinoid receptor 1-dependent withdrawal. Biol Psychiatry. 2015;77:475–87.

    Article  CAS  PubMed  Google Scholar 

  78. Rahn EJ, Deng L, Thakur GA, Vemuri K, Zvonok AM, Lai YY, et al. Prophylactic cannabinoid administration blocks the development of paclitaxel-induced neuropathic nociception during analgesic treatment and following cessation of drug delivery. Mol Pain. 2014;10:27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Pascual D, Goicoechea C, Suardiaz M, Martin MI. A cannabinoid agonist, WIN 55,212-2, reduces neuropathic nociception induced by paclitaxel in rats. Pain. 2005;118:23–34.

    Article  CAS  PubMed  Google Scholar 

  80. Guindon J, Lai Y, Takacs SM, Bradshaw HB, Hohmann AG. Alterations in endocannabinoid tone following chemotherapy-induced peripheral neuropathy: effects of endocannabinoid deactivation inhibitors targeting fatty-acid amide hydrolase and monoacylglycerol lipase in comparison to reference analgesics following cisplatin treatment. Pharmacol Res. 2013;67:94–109.

    Article  CAS  PubMed  Google Scholar 

  81. Grim TW, Ghosh S, Hsu KL, Cravatt BF, Kinsey SG, Lichtman AH. Combined inhibition of FAAH and COX produces enhanced anti-allodynic effects in mouse neuropathic and inflammatory pain models. Pharmacol Biochem Behav. 2014;124:405–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Long JZ, Nomura DK, Vann RE, Walentiny DM, Booker L, Jin X, et al. Dual blockade of FAAH and MAGL identifies behavioral processes regulated by endocannabinoid crosstalk in vivo. Proc Natl Acad Sci USA. 2009;106:20270–5.

    Article  PubMed  Google Scholar 

  83. Adamson Barnes NS, Mitchell VA, Kazantzis NP, Vaughan CW. Actions of the dual FAAH/MAGL inhibitor JZL195 in a murine neuropathic pain model. Br J Pharmacol. 2016;173:77–87.

    Article  CAS  PubMed  Google Scholar 

  84. King KM, Myers AM, Soroka-Monzo AJ, Tuma RF, Tallarida RJ, Walker EA, et al. Single and combined effects of Delta(9)-tetrahydrocannabinol and cannabidiol in a mouse model of chemotherapy-induced neuropathic pain. Br J Pharmacol. 2017;174:2832–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Guindon J, Hohmann AG. Use of sodium bicarbonate to promote weight gain, maintain body temperature, normalize renal funtisons and minimize mortality in rodents receiving the chemo therapeutic agent cisplatin. Neursoci. Lett. 2013;544:41–6.

    Article  CAS  Google Scholar 

  86. Cliffer KD, Siuciak JA, Carson SR, Radley HE, Park JS, Lewis DR, et al. Physiological characterization of taxol-induced large-fiber sensory neuropathy in the rat. Ann Neurol. 1998;43:46–55.

    Article  CAS  PubMed  Google Scholar 

  87. Ali BH, Ramkumar A, Madanagopal TT, Waly MI, Tageldin M, Al-Abri S, et al. Motor and behavioral changes in mice with cisplatin-induced acute renal failure. Physiol Res. 2014;63:35–45.

    CAS  PubMed  Google Scholar 

  88. Harris HM, Sufka KJ, Gul W, ElSohly MA. Effects of delta-9-tetrahydrocannabinol and cannabidiol on cisplatin-induced neuropathy in mice. Planta Med. 2016;82:1169–72.

    Article  CAS  PubMed  Google Scholar 

  89. Mulpuri Y, Marty VN, Munier JJ, Mackie K, Schmidt BL, Seltzman HH, et al. Synthetic peripherally-restricted cannabinoid suppresses chemotherapy-induced peripheral neuropathy pain symptoms by CB1 receptor activation. Neuropharmacology. 2018;139:85–97.

    Article  CAS  PubMed  Google Scholar 

  90. Deng L, Guindon J, Vemuri VK, Thakur GA, White FA, Makriyannis A, et al. The maintenance of cisplatin- and paclitaxel-induced mechanical and cold allodynia is suppressed by cannabinoid CB(2) receptor activation and independent of CXCR90 signaling in models of chemotherapy-induced peripheral neuropathy. Mol Pain. 2012;8:71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Vera G, Chiarlone A, Cabezos PA, Pascual D, Martin MI, Abalo R. WIN 55,212-2 prevents mechanical allodynia but not alterations in feeding behaviour induced by chronic cisplatin in the rat. Life Sci. 2007;81:468–79.

    Article  CAS  PubMed  Google Scholar 

  92. Nealon CM, Henderson-Redmond AN, Hale DE, Morgan DJ. Tolerance to WIN55212-2 is delayed in desensitization-resistant S426A/S430A mice. Neuropharmacology. 2019;148:151–9.

    Article  CAS  PubMed  Google Scholar 

  93. Khasabova IA, Khasabov S, Paz J, Harding-Rose C, Simone DA, Seybold VS. Cannabinoid type-1 receptor reduces pain and neurotoxicity produced by chemotherapy. J Neurosci. 2012;32:7091–101.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Khasabova IA, Yao X, Paz J, Lewandowski CT, Lindberg AE, Coicou L, et al. JZL184 is anti-hyperalgesic in a murine model of cisplatin-induced peripheral neuropathy. Pharmacol Res. 2014;90:67–75.

    Article  CAS  PubMed  Google Scholar 

  95. Uhelski ML, Khasabova IA, Simone DA. Inhibition of anandamide hydrolysis attenuates nociceptor sensitization in a murine model of chemotherapy-induced peripheral neuropathy. J Neurophysiol. 2015;113:1501–10.

    Article  CAS  PubMed  Google Scholar 

  96. Ward SJ, Ramirez MD, Neelakantan H, Walker EA. Cannabidiol prevents the development of cold and mechanical allodynia in paclitaxel-treated female C57Bl6 mice. Anesth Analg. 2011;113:947–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Ward SJ, McAllister SD, Kawamura R, Murase R, Neelakantan H, Walker EA. Cannabidiol inhibits paclitaxel-induced neuropathic pain through 5-HT(1A) receptors without diminishing nervous system function or chemotherapy efficacy. Br J Pharmacol. 2014;171:636–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Rahn EJ, Zvonok AM, Thakur GA, Khanolkar AD, Makriyannis A, Hohmann AG. Selective activation of cannabinoid CB2 receptors suppresses neuropathic nociception induced by treatment with the chemotherapeutic agent paclitaxel in rats. J Pharmacol Exp Ther. 2008;327:584–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Deng L, Cornett BL, Mackie K, Hohmann AG. CB1 knockout mice unveil sustained CB2- mediated antiallodynic effects of the mixed CB1/CB2 agonist CP55,940 in a mouse model of paclitaxel-induced neuropathic pain. Mol Pharmacol. 2015;88:64–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Naguib M, Xu JJ, Diaz P, Brown DL, Cogdell D, Bie B, et al. Prevention of paclitaxel-induced neuropathy through activation of the central cannabinoid type 2 receptor system. Anesth Analg. 2012;114:1104–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Wu J, Hocevar M, Bie B, Foss JF, Naguib M. Cannabinoid type 2 receptor system modulates paclitaxel-induced microglial dysregulation and central sensitization in rats. J Pain. 2018. https://doi.org/10.1016/j.jpain.2018.10.007.

    Article  PubMed  Google Scholar 

  102. Xu JJ, Diaz P, Astruc-Diaz F, Craig S, Munoz E, Naguib M. Pharmacological characterization of a novel cannabinoid ligand, MDA19, for treatment of neuropathic pain. Anesth Analg. 2010;111:99–109.

    Article  PubMed  PubMed Central  Google Scholar 

  103. Segat GC, Manjavachi MN, Matias DO, Passos GF, Freitas CS, Costa R, et al. Antiallodynic effect of beta-caryophyllene on paclitaxel-induced peripheral neuropathy in mice. Neuropharmacology. 2017;125:207–19.

    Article  CAS  PubMed  Google Scholar 

  104. Curry ZA, Wilkerson JL, Bagdas D, Kyte SL, Patel N, Donvito G, et al. Monoacylglycerol lipase inhibitors reverse paclitaxel-induced nociceptive behavior and proinflammatory markers in a mouse model of chemotherapy-induced neuropathy. J Pharmacol Exp Ther. 2018;366:169–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Rahn EJ, Makriyannis A, Hohmann AG. Activation of cannabinoid CB1 and CB2 receptors suppresses neuropathic nociception evoked by the chemotherapeutic agent vincristine in rats. Br J Pharmacol. 2007;152:765–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Caprioli A, Coccurello R, Rapino C, Di Serio S, Di Tommaso M, Vertechy M, et al. The novel reversible fatty acid amide hydrolase inhibitor ST4070 increases endocannabinoid brain levels and counteracts neuropathic pain in different animal models. J Pharmacol Exp Ther. 2012;342:188–95.

    Article  CAS  PubMed  Google Scholar 

  107. Britch SC, Wiley JL, Yu Z, Clowers BH, Craft RM. Cannabidiol-Δ9-tetrahydrocannabinol interactions on acute pain and locomotor activity. Drug Alcohol Depend. 2017;175:187–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Meng H, Johnston B, Englesaskis M, Moulin DE, Bhatia A. Selective cannabinoids for chronic neuropathic pain: a systematic review and meta-analysis. Anesth Analg. 2017;125:1638–52.

    Article  CAS  PubMed  Google Scholar 

  109. Nugent SM, Morasco BJ, O’Neil ME, Freeman M, Low A, Kondo K, et al. The effects of cannabis among adults with chronic pain and an overview of general harms: a systemic review. Ann Intern Med. 2017;167:319–31.

    Article  PubMed  Google Scholar 

  110. Andreae MH, Carter GM, Shaparin N, Suslov K, Ellis RJ, Ware MA, et al. Inhaled cannabis for chronic neuropathic pain: a meta-analysis of individual patient data. J Pain. 2015;16:1221–32.

    Article  PubMed  PubMed Central  Google Scholar 

  111. Stockings E, Campbell G, Hall WD, Nielsen S, Zagic D, Rahman R, et al. Cannabis and cannabinoids for the treatment of people with chronic noncancer pain conditions: a systematic review and meta-analysis of controlled and observational studies. Pain. 2018;159:1932–54.

    Article  CAS  PubMed  Google Scholar 

  112. Wilsey B, Marcotte T, Tsodikov A, Millman J, Bentley H, Gouaux B, et al. A randomized, placebo-controlled, crossover trial of cannabis cigarettes in neuropathic pain. J Pain. 2008;9:506–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Lynch ME, Cesar-Rittenberg P, Hohmann AG. A double-blind, placebo-controlled, crossover pilot trial with extension using an oral mucosal cannabinoid extract for treatment of chemotherapy-induced neuropathic pain. J Pain Symptom Manag. 2014;47:166–73.

    Article  Google Scholar 

  114. Serpell M, Ratcliffe S, Hovorka J, Schofield M, Taylor L, Lauder H, et al. A double-blind, randomized, placebo-controlled, parallel group study of THC/CBD spray in peripheral neuropathic pain treatment. Eur J Pain. 2014;18:999–1012.

    Article  CAS  PubMed  Google Scholar 

  115. Nurmikko TJ, Serpell MG, Hoggart B, Toomey PJ, Morlion BJ, Haines D. Sativex successfully treats neuropathic pain characterised by allodynia: a randomised, double-blind, lacebo-controlled clinical trial. Pain. 2007;133:210–20.

    Article  CAS  PubMed  Google Scholar 

  116. Wade DT, Robson P, House H, Makela P, Aram J. A preliminary controlled study to determine whether whole-plant cannabis extracts can improve intractable neurogenic symptoms. Clin Rehabil. 2003;17:21–9.

    Article  PubMed  Google Scholar 

  117. Frank B, Serpell MG, Hughes J, Matthews JN, Kapur D. Comparison of analgesic effects and patient tolerability of nabilone and dihydrocodeine for chronic neuropathic pain: randomised, crossover, double blind study. BMJ. 2008;336:199–201.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. 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.

    Article  CAS  PubMed  Google Scholar 

  119. 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;329:253.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Turcotte D, Doupe M, Torabi M, Gomori A, Ethans K, Esfahani F, et al. Nabilone as an adjunctive to gabapentin for multiple sclerosis-induced neuropathic pain: a randomized controlled trial. Pain Med. 2015;16:149–59.

    Article  PubMed  Google Scholar 

  121. 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.

    Article  CAS  PubMed  Google Scholar 

  122. 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 Delta9-tetrahydrocannabinol in patients with progressive multiple sclerosis. Clin Ther. 2018;40:1467–82.

    Article  CAS  PubMed  Google Scholar 

  123. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Rintala DH, Fiess RN, Tan G, Holmes SA, Bruel BM. Effect of dronabinol on central neuropathic pain after spinal cord injury: a pilot study. Am J Phys Med Rehabil. 2010;89:840–8.

    Article  PubMed  Google Scholar 

  125. Wilsey BL, Deutsch R, Samara E, Marcotte TD, Barnes AJ, Huestis MA, et al. A preliminary evaluation of the relationship of cannabinoid blood concentrations with the analgesic response to vaporized cannabis. J Pain Res. 2016;9:587–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Ware MA, Wang T, Shapiro S, Robinson A, Ducruet T, Huynh T, Gamsa A, Bennett GJ, Collet JP. Smoked cannabis for chronic neuropathic pain: a randomized controlled trial. CMAJ. 2010;182:E694–701.

    Article  PubMed  PubMed Central  Google Scholar 

  127. Wilsey B, Marcotte T, Deutsch R, Gouaux B, Sakai S, Donaghe H. Low-dose vaporized cannabis significantly improves neuropathic pain. J Pain. 2013;14:136–48.

    Article  CAS  PubMed  Google Scholar 

  128. 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. 2017;68:515–21.

    Article  Google Scholar 

  129. Ellis RJ, Toperoff W, Vaida F, van den Brande G, Gonzales J, Gouaux B, et al. Smoked cannabis for neuropathic pain in HIV: a randomized, crossover clinical trial. Neuropsychopharmacology. 2009;34:672–80.

    Article  CAS  PubMed  Google Scholar 

  130. Benedetti F, Carlino E, Pollo A. How placebos change the patient’s brain. Neuropsychopharmacology. 2011;36:339–54.

    Article  PubMed  Google Scholar 

  131. Tetreault P, Mansour A, Vachon-Presseau E, Schnitzer TJ, Apkarian AV, Baliki MN. Brain connectivity predicts placebo response across chronic pain clinical trials. PLoS Biol. 2016;14:e1002570.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Gertsch J. The intricate influence of the placebo effect on medical cannabis and cannabinoids. Med Cannabis Cannabinoids. 2018;1:60–4.

    Article  Google Scholar 

  133. Benedetti F, Amanzio M, Rosato R, Blanchard C. Nonopioid placebo analgesia is mediated by CB1 receptors. Nat Med. 2011;17:1228–30.

    Article  CAS  PubMed  Google Scholar 

  134. Deadwyler SA, Hampson RE, Mu J, Whyte A, Childers S. Cannabinoids modulate voltage sensitive potassium A-current in hippocampal neurons via a cAMP-dependent process. J Pharmacol Exp Ther. 1995;273:734–43.

    CAS  PubMed  Google Scholar 

  135. Shen M, Thayer SA. Cannabinoid receptor agonists protect cultured rat hippocampal neurons from excitotoxicity. Mol Pharmacol. 1998;54(3):459–62.

    Article  CAS  PubMed  Google Scholar 

  136. Ross HR, Napier I, Connor M. Inhibition of recombinant human T-type calcium channels by Delta9-tetrahydrocannabinol and cannabidiol. J Biol Chem. 2008;283:16124–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Bourinet E, Francois A, Laffray S. T-type calcium channels in neuropathic pain. Pain. 2016;157(Suppl 1):S15–22.

    Article  PubMed  Google Scholar 

  138. Caterina MJ, Schumacher MA, Tominaga M, Rosen TA, Levine JD, Julius D. The capsaicin receptor: a heat-activated ion channel in the pain pathway. Nature. 1997;389:816–24.

    Article  CAS  PubMed  Google Scholar 

  139. Holzer P. Capsaicin: cellular targets, mechanisms of action, and selectivity for thin sensory neurons. Pharmacol Rev. 1991;43:143–201.

    CAS  PubMed  Google Scholar 

  140. Lizanecz E, Bagi Z, Pasztor ET, Papp Z, Edes I, Kedei N, et al. Phosphorylation-dependent desensitization by anandamide of vanilloid receptor-1 (TRPV1) function in rat skeletal muscle arterioles and in Chinese hamster ovary cells expressing TRPV1. Mol Pharmacol. 2006;69:1015–23.

    Article  CAS  PubMed  Google Scholar 

  141. Bisogno T, Hanus L, De Petrocellis L, Tchilibon S, Ponde D, Brandi I, et al. Molecular targets for cannabidiol and its synthetic analogues: effect on vanilloid VR1 receptors and on the cellular uptake and enzymatic hydrolysis of anandamide. Br J Pharmacol. 2001;134:845–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. De Gregorio D, McLaughlin RJ, Posa L, Ochoa-Sanchez R, Enns J, Lopez-Canul M, et al. Cannabidiol modulates serotonergic transmission and reverses both allodynia and anxiety-like behavior in a model of neuropathic pain. Pain. 2019;160:136–50.

    Article  CAS  PubMed  Google Scholar 

  143. De Petrocellis L, Vellani V, Schiano-Moriello A, Marini P, Magherini PC, Orlando P, et al. Plant-derived cannabinoids modulate the activity of transient receptor potential channels of ankyrin type-1 and melastatin type-8. J Pharmacol Exp Ther. 2008;325:1007–15.

    Article  CAS  PubMed  Google Scholar 

  144. Hejazi N, Zhou C, Oz M, Sun H, Ye JH, Zhang L. Delta9-tetrahydrocannabinol andendogenous cannabinoid anandamide directly potentiate the function of glycine receptors. Mol Pharmacol. 2006;69:991–7.

    Article  CAS  PubMed  Google Scholar 

  145. Ahrens J, Demir R, Leuwer M, de la Roche J, Krampfl K, Foadi N, et al. The nonpsychotropic cannabinoid cannabidiol modulates and directly activates alpha-1 and alpha-1-Beta glycine receptor function. Pharmacology. 2009;83:217–22.

    Article  CAS  PubMed  Google Scholar 

  146. Xiong W, Cheng K, Cui T, Godlewski G, Rice KC, Xu Y, et al. Cannabinoid potentiation of glycine receptors contributes to cannabis-induced analgesia. Nat Chem Biol. 2011;7:296–303.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Xiong W, Cui T, Cheng K, Yang F, Chen SR, Willenbring D, et al. Cannabinoids suppress inflammatory and neuropathic pain by targeting alpha3 glycine receptors. J Exp Med. 2012;209:1121–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Lau BK, Vaughan CW. Descending modulation of pain: the GABA disinhibition hypothesis of analgesia. Curr Opin Neurobiol. 2014;29:159–64.

    Article  CAS  PubMed  Google Scholar 

  149. Palazzo E, Marabese I, de Novellis V, Oliva P, Rossi F, Berrino L, et al. Metabotropic and NMDA glutamate receptors participate in the cannabinoid-induced antinociception. Neuropharmacology. 2001;40:319–26.

    Article  CAS  PubMed  Google Scholar 

  150. de Novellis V, Mariani L, Palazzo E, Vita D, Marabese I, Scafuro M, et al. Periaqueductal grey CB1 cannabinoid and metabotropic glutamate subtype 5 receptors modulate changes in rostral ventromedial medulla neuronal activities induced by subcutaneous formalin in the rat. Neuroscience. 2005;134:269–81.

    Article  CAS  PubMed  Google Scholar 

  151. Palazzo E, Luongo L, Bellini G, Guida F, Marabese I, Boccella S, et al. Changes in cannabinoid receptor subtype 1 activity and interaction with metabotropic glutamate subtype 5 receptors in the periaqueductal gray-rostral ventromedial medulla pathway in a rodent neuropathic pain model. CNS Neurol Disord Drug Targets. 2012;11:148–61.

    Article  CAS  PubMed  Google Scholar 

  152. Ghalandari-Shamami M, Hassanpour-Ezatti M, Haghparast A. Intra-accumbal NMDA but not AMPA/kainate receptor antagonist attenuates WIN55,212-2 cannabinoid receptor agonist-induced antinociception in the basolateral amygdala in a rat model of acute pain. Pharmacol Biochem Behav. 2011;100:213–9.

    Article  CAS  PubMed  Google Scholar 

  153. Ji G, Neugebauer V. CB1 augments mGluR5 function in medial prefrontal cortical neurons to inhibit amygdala hyperactivity in an arthritis pain model. Eur J Neurosci. 2014;39:455–66.

    Article  PubMed  PubMed Central  Google Scholar 

  154. Kiritoshi T, Ji G, Neugebauer V. Rescue of impaired mGluR5-driven endocannabinoid signaling restores prefrontal cortical output to inhibit pain in arthritic rats. J Neurosci. 2016;36:837–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Seyrek M, Kahraman S, Deveci MS, Yesilyurt O, Dogrul A. Systemic cannabinoids produce CB(1)-mediated antinociception by activation of descending serotonergic pathways that act upon spinal 5-HT(7) and 5-HT(2A) receptors. Eur J Pharmacol. 2010;649(1–3):183–94.

    Article  CAS  PubMed  Google Scholar 

  156. Meng ID, Manning BH, Martin WJ, Fields HL. An analgesia circuit activated by cannabinoids. Nature. 1998;395:381–3.

    Article  CAS  PubMed  Google Scholar 

  157. Bambico FR, Katz N, Debonnel G, Gobbi G. Cannabinoids elicit antidepressant-like behavior and activate serotonergic neurons through the medial prefrontal cortex. J Neurosci. 2007;27:11700–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Yang KH, Isaev D, Morales M, Petroianu G, Galadari S, Oz M. The effect of Delta9-tetrahydrocannabinol on 5-HT3 receptors depends on the current density. Neuroscience. 2010;171:40–9.

    Article  CAS  PubMed  Google Scholar 

  159. Pertwee RG. Cannabinoid pharmacology: the first 66 years. Br J Pharmacol. 2006;147:S163–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Eljaschewitsch E, Witting A, Mawrin C, Lee T, Schmidt PM, Wolf S, et al. The endocannabinoid anandamide protects neurons during CNS inflammation by induction of MKP-1 in microglial cells. Neuron. 2006;49:67–79.

    Article  CAS  PubMed  Google Scholar 

  161. Jean-Gilles L, Braitch M, Latif ML, Aram J, Fahey AJ, Edwards LJ, et al. Effects of pro-inflammatory cytokines on cannabinoid CB1 and CB2 receptors in immune cells. Acta Physiol (Oxf). 2015;214:63–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Kozela E, Pietr M, Juknat A, Rimmerman N, Levy R, Vogel Z. Cannabinoids Delta(9)-tetrahydrocannabinol and cannabidiol differentially inhibit the lipopolysaccharide-activated NF-kappaB and interferon-beta/STAT proinflammatory pathways in BV-2 microglial cells. J Biol Chem. 2010;285:1616–26.

    Article  CAS  PubMed  Google Scholar 

  163. Malek N, Popiolek-Barczyk K, Mika J, Przewlocka B, Starowicz K. Anandamide, acting via CB2 receptors, alleviates LPS-induced neuroinflammation in rat primary microglial cultures. Neural Plast. 2015;2015:130639.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Katona S, Kaminski E, Sanders H, Zajicek J. Cannabinoid influence on cytokine profile in multiple sclerosis. Clin Exp Immunol. 2005;140:580–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Coopman K, Smith LD, Wright KL, Ward SG. Temporal variation in CB2R levels following T lymphocyte activation: evidence that cannabinoids modulate CXCL12-induced chemotaxis. Int Immunopharmacol. 2007;7:360–71.

    Article  CAS  PubMed  Google Scholar 

  166. Ghosh S, Preet A, Groopman JE, Ganju RK. Cannabinoid receptor CB2 modulates the CXCL12/CXCR166-mediated chemotaxis of T lymphocytes. Mol Immunol. 2006;43:2169–79.

    Article  CAS  PubMed  Google Scholar 

  167. Lombard C, Nagarkatti M, Nagarkatti P. CB2 cannabinoid receptor agonist, JWH-015, triggers apoptosis in immune cells: potential role for CB2-selective ligands as immunosuppressive agents. Clin Immunol. 2007;122:259–70.

    Article  CAS  PubMed  Google Scholar 

  168. Desai PB, Duan JZ, Zhu YW, Kouzi S. Human liver microsomal metabolism of paclitaxel and drug interactions. Eur J Drug Metab Pharmacokinet. 1998;23:417–24.

    Article  CAS  PubMed  Google Scholar 

  169. Spratlin J, Sawyer MB. Pharmacogenetics of paclitaxel metabolism. Crit Rev Oncol Hematol. 2007;61:222–9.

    Article  PubMed  Google Scholar 

  170. Engels FK, Ten Tije AJ, Baker SD, Lee CK, Loos WJ, Vulto AG, et al. Effect of cytochrome P450 3A4 inhibition on the pharmacokinetics of docetaxel. Clin Pharmacol Ther. 2004;75:448–54.

    Article  CAS  PubMed  Google Scholar 

  171. Dennison JB, Kulanthaivel P, Barbuch RJ, Renbarger JL, Ehlhardt WJ, Hall SD. Selective metabolism of vincristine in vitro by CYP3A5. DMD. 2006;34:1317–27.

    Article  CAS  Google Scholar 

  172. Zendulka O, Dovrtelova G, Noskova K, Turjap M, Sulcova A, Hanus L, et al. Cannabinoids and cytochrome P450 interactions. Curr Drug Metab. 2016;17:206–26.

    Article  CAS  PubMed  Google Scholar 

  173. Bergamaschi MM, Queiroz RH, Chagas MH, de Oliveira DC, De Martinis BS, Kapczinski F, et al. Cannabidiol reduces the anxiety induced by simulated public speaking in treatment-naive social phobia patients. Neuropsychopharmacology. 2011;36:1219–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Yamaori S, Ebisawa J, Okushima Y, Yamamoto I, Watanabe K. Potent inhibition of human cytochrome P450 3A isoforms by cannabidiol: role of phenolic hydroxyl groups in the resorcinol moiety. Life Sci. 2011;88:730–6.

    Article  CAS  PubMed  Google Scholar 

  175. Ohlsson A, Lindgren JE, Andersson S, Agurell S, Gillespie H, Hollister LE. Single-dose kinetics of deuterium-labelled cannabidiol in man after smoking and intravenous administration. Biomed Environ Mass Spectrom. 1986;13:77–83.

    Article  CAS  PubMed  Google Scholar 

  176. White CM. A review of human studies assessing cannabidiol’s (CBD) therapeutic actions and potential. J Clin Pharmacol. 2019. https://doi.org/10.1002/jcph.1387.

    Article  PubMed  Google Scholar 

  177. Ghozland S, Matthes HW, Simonin F, Filliol D, Kieffer BL, Maldonado R. Motivational effects of cannabinoids are mediated by mu-opioid and kappa-opioid receptors. J Neurosci. 2002;22:1146–54.

    Article  CAS  PubMed  Google Scholar 

  178. Valverde O, Ledent C, Beslot F, Parmentier M, Roques BP. Reduction of stress-induced analgesia but not of exogenous opioid effects in mice lacking CB1 receptors. Eur J Neurosci. 2000;12:533–9.

    Article  CAS  PubMed  Google Scholar 

  179. Welch SP. Blockade of cannabinoid-induced antinociception by norbinaltorphimine, but not N,N-diallyl-tyrosine-Aib-phenylalanine-leucine, ICI 174,864 or naloxone in mice. J Pharmacol Exp Ther. 1993;265:633–40.

    CAS  PubMed  Google Scholar 

  180. da Fonseca Pacheco D, Klein A, Perez AC, da Fonseca Pacheco CM, de Francischi N, Reis GML, Duarte IDG. Central antinociception induced by µ-opioid receptor agonist morphine, but not δ- or κ-, is mediated by cannabinoid CB1 receptor. BJP. 2009;158:225–31.

    Article  CAS  Google Scholar 

  181. da Fonseca Pacheco D, Klein A, de Castro Perez A, da Fonseca Pacheco CM, de Francischi JN, Duarte ID. The mu-opioid receptor agonist morphine, but not agonists at delta- or kappa-opioid receptors, induces peripheral antinociception mediated by cannabinoid receptors. Br J Pharmacol. 2008;154:1143–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Ibrahim MM, Porecca F, Lai J, Albrecht PJ, Rice FL, Khodorova A, et al. CB2 cannabinoid receptor activation produces antinociception by stimulating peripheral release of endogenous opioids. PNAS. 2004;102:3093–8.

    Article  CAS  Google Scholar 

  183. Curto-Reyes V, Boto T, Hidalgo A, Menendez L, Baamonde A. Antinociceptive effects induced through the stimulation of spinal cannabinoid type 2 receptors in chronically inflamed mice. Eur J Pharmacol. 2011;668:184–9.

    Article  CAS  PubMed  Google Scholar 

  184. Bachhuber MA, Saloner B, Cunningham CO, Feingold J, Barry CL. Could Delaware’s medical marijuana law reduce harms from opioid analgesics? Del Med J. 2014;86:341–3.

    PubMed  Google Scholar 

  185. Abrams DI, Couey P, Shade SB, Kelly ME, Benowitz NL. Cannabinoid–opioid interaction in chronic pain. Clin Pharmacol Ther. 2011;90:844–51.

    Article  CAS  PubMed  Google Scholar 

  186. Johnson JR, Lossignol D, Burnell-Nugent M, Fallon MT. An open-label extension study to investigate the long-term safety and tolerability of THC/CBD oromucosal spray and oromucosal THC spray in patients with terminal cancer-related pain refractory to strong opioid analgesics. J Pain Symptom Manag. 2013;46:207–18.

    Article  CAS  Google Scholar 

  187. Narang S, Gibson D, Wasan AD, Ross EL, Michna E, Nedeljkovic SS, et al. Efficacy ofdronabinol as an adjuvant treatment for chronic pain patients on opioid therapy. J Pain. 2008;9:254–64.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work is dedicated to my beloved mother, Manon Marcotte, who passed away from lymphoma on January 3, 2011 (JG), and all the people who have lost their battles against cancer and to all those who are still fighting this dreadful disease. Special acknowledgement is given to Terri Lloyd, who is currently fighting triple-negative breast cancer and is an Admissions Director for the Graduate School of Biomedical Sciences at Texas Tech University Health Sciences Center.

Funding

Funding

This work has been supported by the National Institute on Drug Abuse (DA044999-01A1) (DJM and JG), the Texas Tech University Health Sciences Center School of Medicine (Grant 121035) (JG), the CH Foundation (MN), and the Cancer Prevention and Research Institute of Texas (RP140008) (KP).

Conflict of interest

The authors, Henry L. Blanton, Jennifer Brelsfoard, Nathan DeTurk, Kevin Pruitt, Madhusudhanan Narasimhan, Daniel J. Morgan and Josée Guindon, declare that they have no competing interests.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Josée Guindon.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Blanton, H.L., Brelsfoard, J., DeTurk, N. et al. Cannabinoids: Current and Future Options to Treat Chronic and Chemotherapy-Induced Neuropathic Pain. Drugs 79, 969–995 (2019). https://doi.org/10.1007/s40265-019-01132-x

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40265-019-01132-x

Navigation