Skip to main content

Advertisement

Log in

A Review of Recent Advances in the Therapeutic Uses of Secondary Cannabinoids

  • Cannabis (F Filbey, Section Editor)
  • Published:
Current Addiction Reports Aims and scope Submit manuscript

Abstract

Research supporting the therapeutic efficacy of cannabis remains scant, with the majority of empirical work having focused on the therapeutic potential of Δ9-tetrahydrocannabinol (THC). The aim of the present review is to document recent advances in the study of “secondary” (non-THC) cannabinoids so as to highlight promising avenues for continued research. A number of reviewed studies demonstrate the therapeutic potential of cannabidiol (CBD) for anxiety, depression, pain, psychosis, and epilepsy. Other work has documented the effects of CBD in combination with THC, showing efficacy in the treatment of multiple sclerosis-related spasticity, with mixed evidence for the treatment of neuropathic pain. Though less comprehensively studied, emerging research has also demonstrated the therapeutic potential of other cannabinoids in terms of anticonvulsant effects, reduced inflammation, neuroprotective effects, and improvement of insulin sensitivity. Evidence suggests that further and more nuanced research on cannabinoids is paramount, particularly for CBD and other “secondary” cannabinoids.

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.

Institutional subscriptions

Similar content being viewed by others

References

Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance

  1. Belendiuk KA, Baldini LL, Bonn-Miller MO. Narrative review of the safety and efficacy of marijuana for the treatment of commonly state-approved medical and psychiatric disorders. Addict Sci Clin Pract. 2015;10(1):10.

    Article  PubMed  PubMed Central  Google Scholar 

  2. ProCon.org (US). Number of legal medical marijuana patients [Internet]. Santa Monica (CA): ProCon.org. 2015 [cited 2016 Jan 20]. Available from: http://medicalmarijuana.procon.org/view.resource.php?resourceID=005889.

  3. Hill KP. Medical marijuana for treatment of chronic pain and other medical and psychiatric problems: a clinical review. JAMA. 2015;313(24):2474–83.

    Article  CAS  PubMed  Google Scholar 

  4. Whiting PF, Wolff RF, Deshpande S, Di Nisio M, Duffy S, Hernandez AV, et al. Cannabinoids for medical use: a systematic review and meta-analysis. JAMA. 2015;313(24):2456–73.

    Article  CAS  PubMed  Google Scholar 

  5. ElSohly MA, Slade D. Chemical constituents of marijuana: the complex mixture of natural cannabinoids. Life Sci. 2005;78(5):539–48.

    Article  CAS  PubMed  Google Scholar 

  6. Hillig KW, Mahlberg PG. A chemotaxonomic analysis of cannabinoid variation in cannabis (cannabaceae). Am J Bot. 2004;91(6):966–75.

    Article  CAS  PubMed  Google Scholar 

  7. Walter C, Oertel B, Lötsch J. THC may reproducibly induce electrical hyperalgesia in healthy volunteers. Eur J Pain. 2015;19(4):516–8.

    Article  CAS  PubMed  Google Scholar 

  8. Notcutt WG. Clinical use of cannabinoids for symptom control in multiple sclerosis. Neurotherapeutics. 2015;12(4):769–77.

    Article  CAS  PubMed  Google Scholar 

  9. Harris GE, Dupuis L, Mugford GJ, Johnston L, Haase D, Page G, et al. Patterns and correlates of cannabis use among individuals with HIV/AIDS in maritime Canada. Can J Infect Dis Med Microbiol. 2014;25(1):e1.

    PubMed  PubMed Central  Google Scholar 

  10. Abrams DI, Guzman M. Cannabis in cancer care. Clin Pharmacol Ther. 2015;97(6):575–86.

    Article  CAS  PubMed  Google Scholar 

  11. Almeida V, Levin R, Peres FF, Niigaki ST, Calzavara MB, Zuardi AW, et al. Cannabidiol exhibits anxiolytic but not antipsychotic property evaluated in the social interaction test. Prog Neuro-Psychopharmacol Biol Psychiatry. 2013;41:30–5.

    Article  CAS  Google Scholar 

  12. Bergamaschi MM, Queiroz RHC, Chagas MHN, de Oliveira DCG, 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(6):1219–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Bhattacharyya S, Morrison PD, Fusar-Poli P, Martin-Santos R, Borgwardt S, Winton-Brown T, et al. Opposite effects of Δ-9-tetrahydrocannabinol and cannabidiol on human brain function and psychopathology. Neuropsychopharmacology. 2010;35(3):764–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Campos AC, Ferreira FR, Guimaraes FS. Cannabidiol blocks long-lasting behavioral consequences of predator threat stress: possible involvement of 5HT1A receptors. J Psychiatr Res. 2012;46(11):1501–10.

    Article  PubMed  Google Scholar 

  15. Campos AC, de Paula SV, Carvalho MC, Ferreira FR, Vicente MA, Brandão ML, et al. Involvement of serotonin-mediated neurotransmission in the dorsal periaqueductal gray matter on cannabidiol chronic effects in panic-like responses in rats. Psychopharmacology. 2013;226(1):13–24.

    Article  CAS  PubMed  Google Scholar 

  16. Campos AC, Ortega Z, Palazuelos J, Fogaça MV, Aguiar DC, Diaz-Alonso J, et al. The anxiolytic effect of cannabidiol on chronically stressed mice depends on hippocampal neurogenesis: involvement of the endocannabinoid system. Int J Neuropsychopharmacol. 2013;16(6):1407–19.

    Article  CAS  PubMed  Google Scholar 

  17. Chagas M, Eckeli A, Zuardi A, Pena‐Pereira M, Sobreira‐Neto M, Sobreira E, et al. Cannabidiol can improve complex sleep‐related behaviours associated with rapid eye movement sleep behaviour disorder in Parkinson’s disease patients: a case series. J Clin Pharm Ther. 2014;39(5):564–6.

    Article  CAS  PubMed  Google Scholar 

  18. Crippa JAS, Derenusson GN, Ferrari TB, Wichert-Ana L, Duran FL, Martin-Santos R, et al. Neural basis of anxiolytic effects of cannabidiol (CBD) in generalized social anxiety disorder: a preliminary report. J Psychopharmacol. 2011;25(1):121–30.

    Article  CAS  PubMed  Google Scholar 

  19. Das RK, Kamboj SK, Ramadas M, Yogan K, Gupta V, Redman E, et al. Cannabidiol enhances consolidation of explicit fear extinction in humans. Psychopharmacology. 2013;226(4):781–92. Randomized controlled trial that found CBD to help extinction learning in a fear conditioning paradigm. This indicated the potential benefit of CBD in conjunction with extinction therapies for anxiety disorders.

    Article  CAS  PubMed  Google Scholar 

  20. Deiana S, Watanabe A, Yamasaki Y, Amada N, Arthur M, Fleming S, et al. Plasma and brain pharmacokinetic profile of cannabidiol (CBD), cannabidivarine (CBDV), Δ9-tetrahydrocannabivarin (THCV) and cannabigerol (CBG) in rats and mice following oral and intraperitoneal administration and CBD action on obsessive–compulsive behaviour. Psychopharmacology. 2012;219(3):859–73.

    Article  CAS  PubMed  Google Scholar 

  21. Devinsky O, Marsh E, Friedman D, Thiele E, Laux L, Sullivan J, et al. Cannabidiol in patients with treatment-resistant epilepsy: an open-label interventional trial. Lancet Neurol. 2015. doi:10.1016/S1474-4422(15)00379-8. Provides support for the use of isolated cannabidiol as an anticonvulsant in addition to existing anti-epileptic drug treatment regimens in treatment-resistant pediatric epilepsy.

    PubMed  Google Scholar 

  22. Di Forti M, Sallis H, Allegri F, Trotta A, Ferraro L, Stilo SA, et al. Daily use, especially of high-potency cannabis, drives the earlier onset of psychosis in cannabis users. Schizophr Bull. 2014;40(6):1509–17.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Fogaça M, Reis F, Campos A, Guimarães F. Effects of intra-prelimbic prefrontal cortex injection of cannabidiol on anxiety-like behavior: involvement of 5HT 1A receptors and previous stressful experience. Eur Neuropsychopharmacol. 2014;24(3):410–9. Animal study that implicates 5HT1A receptors as a mechanism of treatment efficacy, and previous stressful experience as a moderator for the anxiolytic effects of CBD.

    Article  PubMed  Google Scholar 

  24. Gomes FV, Resstel LB, Guimarães FS. The anxiolytic-like effects of cannabidiol injected into the bed nucleus of the stria terminalis are mediated by 5-HT1A receptors. Psychopharmacology. 2011;213(2–3):465–73.

    Article  CAS  PubMed  Google Scholar 

  25. Granjeiro EM, Gomes FV, Guimaraes FS, Corrêa FM, Resstel LB. Effects of intracisternal administration of cannabidiol on the cardiovascular and behavioral responses to acute restraint stress. Pharmacol Biochem Behav. 2011;99(4):743–8.

    Article  CAS  PubMed  Google Scholar 

  26. Jones NA, Glyn SE, Akiyama S, Hill TD, Hill AJ, Weston SE, et al. Cannabidiol exerts anti-convulsant effects in animal models of temporal lobe and partial seizures. Seizure. 2012;21(5):344–52.

    Article  PubMed  Google Scholar 

  27. Leweke F, Piomelli D, Pahlisch F, Muhl D, Gerth C, Hoyer C, et al. Cannabidiol enhances anandamide signaling and alleviates psychotic symptoms of schizophrenia. Transl Psychiatry. 2012;2(3):e94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Liput DJ, Hammell DC, Stinchcomb AL, Nixon K. Transdermal delivery of cannabidiol attenuates binge alcohol-induced neurodegeneration in a rodent model of an alcohol use disorder. Pharmacol Biochem Behav. 2013;111:120–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Long LE, Chesworth R, Huang X-F, McGregor IS, Arnold JC, Karl T. A behavioural comparison of acute and chronic Δ9-tetrahydrocannabinol and cannabidiol in C57BL/6JArc mice. Int J Neuropsychopharmacol. 2010;13(7):861–76.

    Article  CAS  PubMed  Google Scholar 

  30. Marinho A, Vila-Verde C, Fogaça M, Guimarães F. Effects of intra-infralimbic prefrontal cortex injections of cannabidiol in the modulation of emotional behaviors in rats: contribution of 5HT 1A receptors and stressful experiences. Behav Brain Res. 2015;286:49–56.

    Article  CAS  PubMed  Google Scholar 

  31. Porter BE, Jacobson C. Report of a parent survey of cannabidiol-enriched cannabis use in pediatric treatment-resistant epilepsy. Epilepsy Behav. 2013;29(3):574–7. Provides support for the use of cannabidiol as an anticonvulsant in pediatric epilepsy.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Reus GZ, Stringari RB, Ribeiro KF, Luft T, Abelaira HM, Fries GR, et al. Administration of cannabidiol and imipramine induces antidepressant‐like effects in the forced swimming test and increases brain‐derived neurotrophic factor levels in the rat amygdala. Acta Neuropsychiatrica. 2011;23(5):241–8.

    Article  PubMed  Google Scholar 

  33. Ward SJ, McAllister SD, Kawamura R, Murase R, Neelakantan H, Walker EA. Cannabidiol inhibits paclitaxel‐induced neuropathic pain through 5‐HT1A receptors without diminishing nervous system function or chemotherapy efficacy. Br J Pharmacol. 2014;171(3):636–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Zanelati T, Biojone C, Moreira F, Guimaraes F, Joca S. Antidepressant‐like effects of cannabidiol in mice: possible involvement of 5‐HT1A receptors. Br J Pharmacol. 2010;159(1):122–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Allsop DJ, Copeland J, Lintzeris N, Dunlop AJ, Montebello M, Sadler C, et al. Nabiximols as an agonist replacement therapy during cannabis withdrawal: a randomized clinical trial. JAMA Psychiatry. 2014;71(3):281–91. Double-blind, randomized clinical trial of 51 cannabis dependent treatment-seeking individuals. After nabiximol administration, individuals reported reduced cannabis withdrawal symptoms and patient retention improved relative to placebo. This shows the potential of nabiximols as a treatment for cannabis use disorder.

    Article  CAS  PubMed  Google Scholar 

  36. 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(5):451–9.

    Article  CAS  PubMed  Google Scholar 

  37. Flachenecker P, Henze T, Zettl UK. Nabiximols (THC/CBD oromucosal spray, Sativex®) in clinical practice-results of a multicenter, non-interventional study (MOVE 2) in patients with multiple sclerosis spasticity. Eur Neurol. 2014;71(5–6):271–9.

    Article  CAS  PubMed  Google Scholar 

  38. Hoggart B, Ratcliffe S, Ehler E, Simpson K, Hovorka J, Lejčko J, et al. A multicentre, open-label, follow-on study to assess the long-term maintenance of effect, tolerance and safety of THC/CBD oromucosal spray in the management of neuropathic pain. J Neurol. 2015;262(1):27–40. Shows decreased pain and improved sleep quality in a large sample (n = 380) of individuals with neuropathic pain over the course of 38 weeks. Provides evidence for the safety of prolonged nabiximol use.

    Article  CAS  PubMed  Google Scholar 

  39. Johnson JR, Burnell-Nugent M, Lossignol D, Ganae-Motan ED, Potts R, Fallon MT. Multicenter, double-blind, randomized, placebo-controlled, parallel-group study of the efficacy, safety, and tolerability of THC: CBD extract and THC extract in patients with intractable cancer-related pain. J Pain Symptom Manag. 2010;39(2):167–79.

    Article  Google Scholar 

  40. 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(2):207–18.

    Article  Google Scholar 

  41. Langford R, 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(4):984–97. doi:10.1007/s00415-012-6739-4.

    Article  CAS  PubMed  Google Scholar 

  42. 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®(nabiximols). Mult Scler J. 2012;18(2):219–28.

    Article  CAS  Google Scholar 

  43. 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®), as add‐on therapy, in subjects with refractory spasticity caused by multiple sclerosis. Eur J Neurol. 2011;18(9):1122–31.

    Article  CAS  PubMed  Google Scholar 

  44. Portenoy RK, Ganae-Motan ED, Allende S, Yanagihara R, Shaiova L, Weinstein S, et al. Nabiximols for opioid-treated cancer patients with poorly-controlled chronic pain: a randomized, placebo-controlled, graded-dose trial. J Pain. 2012;13(5):438–49.

    Article  CAS  PubMed  Google Scholar 

  45. 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(1):285–95. Clinical trial of repeated Sativex use up to 48 doses per day with no psychoses, adverse event trends, or withdrawal symptoms observed. Demonstrates that Sativex is well tolerated, and can provide sustained reduction in MS spasticity symptoms.

    Article  PubMed  Google Scholar 

  46. Schubart CD, Sommer IE, van Gastel WA, Goetgebuer RL, Kahn RS, Boks MP. Cannabis with high cannabidiol content is associated with fewer psychotic experiences. Schizophr Res. 2011;130(1):216–21.

    Article  PubMed  Google Scholar 

  47. Zuardi A, Crippa J, Hallak J, Pinto J, Chagas M, Rodrigues G, et al. Cannabidiol for the treatment of psychosis in Parkinson’s disease. J Psychopharmacol. 2008;23:979–83.

    Article  PubMed  Google Scholar 

  48. Zuardi AW, Morais SL, Guimaraes FS, Mechoulam R. Antipsychotic effect of cannabidiol. J Clin Psychiatry. 1995;56(10):485–6.

    CAS  PubMed  Google Scholar 

  49. Johnson L, O’Malley P, Miech R, Bachman J, Schulenberg J. Monitoring the future national survey results on drug use: 1975–2013: overview. Key findings on adolescent drug use. Ann Arbor: Institute for Social Research, The University of Michigan; 2014.

    Google Scholar 

  50. Cunha JM, Carlini E, Pereira AE, Ramos OL, Pimentel C, Gagliardi R, et al. Chronic administration of cannabidiol to healthy volunteers and epileptic patients. Pharmacology. 1980;21(3):175–85.

    Article  CAS  PubMed  Google Scholar 

  51. Mechoulam R, Carlini E. Toward drugs derived from cannabis. Naturwissenschaften. 1978;65(4):174–9.

    Article  CAS  PubMed  Google Scholar 

  52. Trembly B, Sherman M. Double-blind clinical study of cannabidiol as a secondary anticonvulsant. International Conference on Cannabis and Cannabinoids. Kolympari, Crete; 1990.

  53. Jones N, Hill A, Hill T, Peres I, Hadid R, Wright S, et al. Assessment of the anticonvulsant effects and tolerability profile of cannabidiol: GW pharmaceuticals’ preclinical program (P4. 255). Neurology. 2015;84(14 Supplement):P4. 255. Demonstrates promising anticonvulsant effects of CBD in vitro and in vivo with a favorable tolerability profile, providing support for future clinical investigation.

  54. Gedde M, Maa E. Whole cannabis extract of high concentration cannabidiol may calm seizures in highly refractory pediatric epilepsies. American Epilepsy Society, 67th annual meeting. 2013.

  55. Ames F, Cridland S. Anticonvulsant effect of cannabidiol. S Afr Med J. 1986;69(1):14.

    CAS  PubMed  Google Scholar 

  56. McPartland JM, Russo EB. Cannabis and cannabis extracts: greater than the sum of their parts? J Cannabis Ther. 2001;1(3–4):103–32.

    Article  CAS  Google Scholar 

  57. Feliú A, Moreno‐Martet M, Mecha M, Carrillo‐Salinas F, Lago E, Fernández‐Ruiz J, et al. A Sativex®‐like combination of phytocannabinoids as a disease‐modifying therapy in a viral model of multiple sclerosis. Br J Pharmacol. 2015;172:3579–95.

    Article  PubMed  Google Scholar 

  58. Valdeolivas S, Satta V, Pertwee RG, Fernández-Ruiz J, Sagredo O. Sativex-like combination of phytocannabinoids is neuroprotective in malonate-lesioned rats, an inflammatory model of Huntington’s disease: role of CB1 and CB2 receptors. ACS Chem Neurosci. 2012;3(5):400–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Sativex oromucosal spray—summary of product characteristics (SPC) - (eMC).

  60. Fernández O. Advances in the management of MS spasticity: recent observational studies. Eur Neurol. 2014;72 Suppl 1:12–4.

    Article  PubMed  Google Scholar 

  61. Koppel BS, Brust JC, Fife T, Bronstein J, Youssof S, Gronseth G, et al. Systematic review: efficacy and safety of medical marijuana in selected neurologic disorders report of the guideline development subcommittee of the American academy of neurology. Neurology. 2014;82(17):1556–63.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Leussink VI, Husseini L, Warnke C, Broussalis E, Hartung H-P, Kieseier BC. Symptomatic therapy in multiple sclerosis: the role of cannabinoids in treating spasticity. Ther Adv Neurol Disord. 2012;5(5):255–66.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Rekand T. THC: CBD spray and MS spasticity symptoms: data from latest studies. Eur Neurol. 2014;71 Suppl 1:4–9.

    Article  CAS  PubMed  Google Scholar 

  64. Syed YY, McKeage K, Scott LJ. Delta-9-tetrahydrocannabinol/cannabidiol (Sativex®): a review of its use in patients with moderate to severe spasticity due to multiple sclerosis. Drugs. 2014;74(5):563–78.

    Article  CAS  PubMed  Google Scholar 

  65. Wade DT, Collin C, Stott C, Duncombe P. Meta-analysis of the efficacy and safety of Sativex (nabiximols), on spasticity in people with multiple sclerosis. Mult Scler. 2010;16(6):707–14.

    Article  CAS  PubMed  Google Scholar 

  66. Nurmikko TJ, Serpell MG, Hoggart B, Toomey PJ, Morlion BJ, Haines D. Sativex successfully treats neuropathic pain characterised by allodynia: a randomised, double-blind, placebo-controlled clinical trial. Pain®. 2007;133(1):210–20.

    Article  CAS  Google Scholar 

  67. Snedecor SJ, Sudharshan L, Cappelleri JC, Sadosky A, Mehta S, Botteman M. Systematic review and meta‐analysis of pharmacological therapies for painful diabetic peripheral neuropathy. Pain Pract. 2014;14(2):167–84.

    Article  PubMed  Google Scholar 

  68. Hill A, Mercier M, Hill T, Glyn S, Jones N, Yamasaki Y, et al. Cannabidivarin is anticonvulsant in mouse and rat. Br J Pharmacol. 2012;167(8):1629–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Hill T, Cascio MG, Romano B, Duncan M, Pertwee R, Williams C, et al. Cannabidivarin‐rich cannabis extracts are anticonvulsant in mouse and rat via a CB1 receptor‐independent mechanism. Br J Pharmacol. 2013;170(3):679–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Borrelli F, Fasolino I, Romano B, Capasso R, Maiello F, Coppola D, et al. Beneficial effect of the non-psychotropic plant cannabinoid cannabigerol on experimental inflammatory bowel disease. Biochem Pharmacol. 2013;85(9):1306–16.

    Article  CAS  PubMed  Google Scholar 

  71. Romano B, Borrelli F, Fasolino I, Capasso R, Piscitelli F, Cascio M, et al. The cannabinoid TRPA1 agonist cannabichromene inhibits nitric oxide production in macrophages and ameliorates murine colitis. Br J Pharmacol. 2013;169(1):213–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Granja AG, Carrillo-Salinas F, Pagani A, Gómez-Cañas M, Negri R, Navarrete C, et al. A cannabigerol quinone alleviates neuroinflammation in a chronic model of multiple sclerosis. J NeuroImmune Pharmacol. 2012;7(4):1002–16.

    Article  PubMed  Google Scholar 

  73. Borrelli F, Pagano E, Romano B, Panzera S, Maiello F, Coppola D, et al. Colon carcinogenesis is inhibited by the TRPM8 antagonist cannabigerol, a cannabis-derived non-psychotropic cannabinoid. Carcinogenesis. 2014. doi:10.1093/carcin/bgu205.

    PubMed  Google Scholar 

  74. Wargent E, Zaibi M, Silvestri C, Hislop D, Stocker C, Stott C, et al. The cannabinoid Δ9-tetrahydrocannabivarin (THCV) ameliorates insulin sensitivity in two mouse models of obesity. Nutr Diabetes. 2013;3(5):e68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Garcia C, Palomo‐Garo C, García‐Arencibia M, Ramos J, Pertwee R, Fernández‐Ruiz J. Symptom‐relieving and neuroprotective effects of the phytocannabinoid Δ9‐THCV in animal models of Parkinson’s disease. Br J Pharmacol. 2011;163(7):1495–506.

  76. Russo EB. Taming THC: potential cannabis synergy and phytocannabinoid‐terpenoid entourage effects. Br J Pharmacol. 2011;163(7):1344–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Andréasson S, Engström A, Allebeck P, Rydberg U. Cannabis and schizophrenia a longitudinal study of Swedish conscripts. Lancet. 1987;330(8574):1483–6.

    Article  Google Scholar 

  78. Arseneault L, Cannon M, Poulton R, Murray R, Caspi A, Moffitt TE. Cannabis use in adolescence and risk for adult psychosis: longitudinal prospective study. BMJ. 2002;325(7374):1212–3.

    Article  PubMed  PubMed Central  Google Scholar 

  79. Di Forti M, Iyegbe C, Sallis H, Kolliakou A, Falcone MA, Paparelli A, et al. Confirmation that the AKT1 (rs2494732) genotype influences the risk of psychosis in cannabis users. Biol Psychiatry. 2012;72(10):811–6.

    Article  PubMed  Google Scholar 

  80. Lucas P, Walsh Z, Crosby K, Callaway R, Belle-Isle L, Kay R, et al. Substituting cannabis for prescription drugs, alcohol and other substances among medical cannabis patients: the impact of contextual factors. Drug Alcohol Rev. 2015. doi:10.1111/dar.12323.

    PubMed  Google Scholar 

  81. Reiman A. Cannabis as a substitute for alcohol and other drugs. Harm Reduction J. 2009;6(1):35. doi:10.1186/1477-7517-6-35.

    Article  Google Scholar 

  82. Adamson SJ, Kay-Lambkin FJ, Baker AL, Lewin TJ, Thornton L, Kelly BJ, et al. An improved brief measure of cannabis misuse: the cannabis use disorders identification test—revised (CUDIT-R). Drug Alcohol Depend. 2010;110(1):137–43.

    Article  PubMed  Google Scholar 

  83. Bujarski S, Galang JN, Short NA, Trafton JA, Gifford E, Kimerling R, et al. Cannabis use disorder treatment barriers/facilitators among veterans with PTSD. Psychol Addict Behav. 2016;30:73–81.

    Article  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Marcel O. Bonn-Miller.

Ethics declarations

Conflict of Interest

Danielle Morabito, Peter Soyster, and Shaw Ramey-Wright declare that they have no conflict of interest. Katherine A. Belendiuk reports stock/equity from Shire Pharmaceuticals, outside the submitted work. Marcel O. Bonn-Miller reports personal fees and non-financial support from Tilray (Division of Privateer Holdings), non-financial support from Americans for Safe Access, and personal fees from Zynerba Pharmaceuticals, outside the submitted work.

Human and Animal Rights and Informed Consent

Among cited articles where one of the authors of the current report were authors, local Institutional Review Board approval was obtained and maintained for studies where human (or animal) subjects research was performed.

Additional information

This article is part of the Topical Collection on Cannabis

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Morabito, D., Soyster, P., Ramey-Wright, S. et al. A Review of Recent Advances in the Therapeutic Uses of Secondary Cannabinoids. Curr Addict Rep 3, 230–238 (2016). https://doi.org/10.1007/s40429-016-0096-9

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40429-016-0096-9

Keywords

Navigation