Skip to main content
Top
Published in: The Journal of Headache and Pain 1/2018

Open Access 01-12-2018 | Research article

Patterns of medicinal cannabis use, strain analysis, and substitution effect among patients with migraine, headache, arthritis, and chronic pain in a medicinal cannabis cohort

Authors: Eric P. Baron, Philippe Lucas, Joshua Eades, Olivia Hogue

Published in: The Journal of Headache and Pain | Issue 1/2018

Login to get access

Abstract

Background

Medicinal cannabis registries typically report pain as the most common reason for use. It would be clinically useful to identify patterns of cannabis treatment in migraine and headache, as compared to arthritis and chronic pain, and to analyze preferred cannabis strains, biochemical profiles, and prescription medication substitutions with cannabis.

Methods

Via electronic survey in medicinal cannabis patients with headache, arthritis, and chronic pain, demographics and patterns of cannabis use including methods, frequency, quantity, preferred strains, cannabinoid and terpene profiles, and prescription substitutions were recorded. Cannabis use for migraine among headache patients was assessed via the ID Migraine™ questionnaire, a validated screen used to predict the probability of migraine.

Results

Of 2032 patients, 21 illnesses were treated with cannabis. Pain syndromes accounted for 42.4% (n = 861) overall; chronic pain 29.4% (n = 598;), arthritis 9.3% (n = 188), and headache 3.7% (n = 75;). Across all 21 illnesses, headache was a symptom treated with cannabis in 24.9% (n = 505). These patients were given the ID Migraine™ questionnaire, with 68% (n = 343) giving 3 “Yes” responses, 20% (n = 102) giving 2 “Yes” responses (97% and 93% probability of migraine, respectively). Therefore, 88% (n = 445) of headache patients were treating probable migraine with cannabis. Hybrid strains were most preferred across all pain subtypes, with “OG Shark” the most preferred strain in the ID Migraine™ and headache groups. Many pain patients substituted prescription medications with cannabis (41.2–59.5%), most commonly opiates/opioids (40.5–72.8%). Prescription substitution in headache patients included opiates/opioids (43.4%), anti-depressant/anti-anxiety (39%), NSAIDs (21%), triptans (8.1%), anti-convulsants (7.7%), muscle relaxers (7%), ergots (0.4%).

Conclusions

Chronic pain was the most common reason for cannabis use, consistent with most registries. The majority of headache patients treating with cannabis were positive for migraine. Hybrid strains were preferred in ID Migraine™, headache, and most pain groups, with “OG Shark”, a high THC (Δ9-tetrahydrocannabinol)/THCA (tetrahydrocannabinolic acid), low CBD (cannabidiol)/CBDA (cannabidiolic acid), strain with predominant terpenes β-caryophyllene and β-myrcene, most preferred in the headache and ID Migraine™ groups. This could reflect the potent analgesic, anti-inflammatory, and anti-emetic properties of THC, with anti-inflammatory and analgesic properties of β-caryophyllene and β-myrcene. Opiates/opioids were most commonly substituted with cannabis. Prospective studies are needed, but results may provide early insight into optimizing crossbred cannabis strains, synergistic biochemical profiles, dosing, and patterns of use in the treatment of headache, migraine, and chronic pain syndromes.
Literature
1.
go back to reference Russo E (1998) Cannabis for migraine treatment: the once and future prescription? An historical and scientific review. Pain 76:3–8PubMedCrossRef Russo E (1998) Cannabis for migraine treatment: the once and future prescription? An historical and scientific review. Pain 76:3–8PubMedCrossRef
2.
go back to reference Baron EP (2015) Comprehensive review of medicinal marijuana, cannabinoids, and therapeutic implications in medicine and headache: what a long strange trip it's been …. Headache 55:885–916PubMedCrossRef Baron EP (2015) Comprehensive review of medicinal marijuana, cannabinoids, and therapeutic implications in medicine and headache: what a long strange trip it's been …. Headache 55:885–916PubMedCrossRef
3.
go back to reference Brunner TF (1973) Marijuana in ancient Greece and Rome? The literary evidence. Bull Hist Med 47:344–355PubMed Brunner TF (1973) Marijuana in ancient Greece and Rome? The literary evidence. Bull Hist Med 47:344–355PubMed
4.
go back to reference Kuddus M, Ginawi IAM, Al-Hazimi A (2013) Cannabis sativa: an ancient wild edible plant of India. Emir J Food Agric 25:736–745CrossRef Kuddus M, Ginawi IAM, Al-Hazimi A (2013) Cannabis sativa: an ancient wild edible plant of India. Emir J Food Agric 25:736–745CrossRef
6.
go back to reference Mikuriya TH (1973) Marijuana: medical papers 1839-1972. Medi-Comp Press, Oakland Mikuriya TH (1973) Marijuana: medical papers 1839-1972. Medi-Comp Press, Oakland
7.
go back to reference O'Shaughnessy WB (1843) On the preparations of the Indian hemp, or gunjah (cannabis indica): their effects on the animal system in health, and their utility in the treatment of tetanus and other convulsive diseases. Prov Med J Retrosp Med Sci 5:363–369 O'Shaughnessy WB (1843) On the preparations of the Indian hemp, or gunjah (cannabis indica): their effects on the animal system in health, and their utility in the treatment of tetanus and other convulsive diseases. Prov Med J Retrosp Med Sci 5:363–369
9.
11.
go back to reference Greene R (1872) Cannabis Indica in the treatment of migraine. Practitioner 41:267–270 Greene R (1872) Cannabis Indica in the treatment of migraine. Practitioner 41:267–270
12.
go back to reference Osler W, McCrae T (1915) The principles and practice of medicine. Appleton, New York Osler W, McCrae T (1915) The principles and practice of medicine. Appleton, New York
13.
14.
go back to reference Farlow JW (1889) On the use of belladonna and cannabis Indica by the rectum in gynecological practice. Boston Med Surg J 120:507–509CrossRef Farlow JW (1889) On the use of belladonna and cannabis Indica by the rectum in gynecological practice. Boston Med Surg J 120:507–509CrossRef
15.
go back to reference Reynolds JR (1890) On the therapeutic uses and toxic effects of cannabis Indica. Lancet 135:637–638CrossRef Reynolds JR (1890) On the therapeutic uses and toxic effects of cannabis Indica. Lancet 135:637–638CrossRef
16.
go back to reference Fishbein M (1942) Migraine associated with menstruation. J Am Med Assoc 237:326 Fishbein M (1942) Migraine associated with menstruation. J Am Med Assoc 237:326
17.
go back to reference British Medical Association (1997) Therapeutic uses of Cannabis. Harwood Academic Publishers, Netherlands British Medical Association (1997) Therapeutic uses of Cannabis. Harwood Academic Publishers, Netherlands
18.
go back to reference Koppel BS, Brust JC, Fife T et al (2014) 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 82:1556–1563PubMedPubMedCentralCrossRef Koppel BS, Brust JC, Fife T et al (2014) 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 82:1556–1563PubMedPubMedCentralCrossRef
19.
go back to reference Aggarwal SK (2013) Cannabinergic pain medicine: a concise clinical primer and survey of randomized-controlled trial results. Clin J Pain 29:162–171PubMedCrossRef Aggarwal SK (2013) Cannabinergic pain medicine: a concise clinical primer and survey of randomized-controlled trial results. Clin J Pain 29:162–171PubMedCrossRef
20.
go back to reference Lynch ME, Ware MA (2015) Cannabinoids for the treatment of chronic non-cancer pain: an updated systematic review of randomized controlled trials. J NeuroImmune Pharmacol 10:293–301PubMedCrossRef Lynch ME, Ware MA (2015) Cannabinoids for the treatment of chronic non-cancer pain: an updated systematic review of randomized controlled trials. J NeuroImmune Pharmacol 10:293–301PubMedCrossRef
21.
go back to reference Lynch ME, Campbell F (2011) Cannabinoids for treatment of chronic non-cancer pain; a systematic review of randomized trials. Br J Clin Pharmacol 72:735–744PubMedPubMedCentralCrossRef Lynch ME, Campbell F (2011) Cannabinoids for treatment of chronic non-cancer pain; a systematic review of randomized trials. Br J Clin Pharmacol 72:735–744PubMedPubMedCentralCrossRef
22.
go back to reference Whiting PF, Wolff RF, Deshpande S et al (2015) Cannabinoids for medical use: a systematic review and meta-analysis. JAMA 313:2456–2473PubMedCrossRef Whiting PF, Wolff RF, Deshpande S et al (2015) Cannabinoids for medical use: a systematic review and meta-analysis. JAMA 313:2456–2473PubMedCrossRef
23.
go back to reference Boychuk DG, Goddard G, Mauro G, Orellana MF (2015) The effectiveness of cannabinoids in the management of chronic nonmalignant neuropathic pain: a systematic review. J Oral Facial Pain Headache 29:7–14PubMedCrossRef Boychuk DG, Goddard G, Mauro G, Orellana MF (2015) The effectiveness of cannabinoids in the management of chronic nonmalignant neuropathic pain: a systematic review. J Oral Facial Pain Headache 29:7–14PubMedCrossRef
24.
go back to reference Committee of the Health Effects of Marijuana (2017) An evidence review and research agenda. The health effects of cannabis and cannabinoids. The current state of evidence and recommendations for research. The National Academies Press, Washington, DC Committee of the Health Effects of Marijuana (2017) An evidence review and research agenda. The health effects of cannabis and cannabinoids. The current state of evidence and recommendations for research. The National Academies Press, Washington, DC
25.
go back to reference Moulin D, Boulanger A, Clark AJ et al (2014) Pharmacological management of chronic neuropathic pain: revised consensus statement from the Canadian Pain Society. Pain Res Manag 19:328–335PubMedPubMedCentralCrossRef Moulin D, Boulanger A, Clark AJ et al (2014) Pharmacological management of chronic neuropathic pain: revised consensus statement from the Canadian Pain Society. Pain Res Manag 19:328–335PubMedPubMedCentralCrossRef
26.
go back to reference Gurley RJ, Aranow R, Katz M (1998) Medicinal marijuana: a comprehensive review. J Psychoactive Drugs 30:137–147PubMedCrossRef Gurley RJ, Aranow R, Katz M (1998) Medicinal marijuana: a comprehensive review. J Psychoactive Drugs 30:137–147PubMedCrossRef
27.
28.
go back to reference Baron EP (2018) Medicinal properties of cannabinoids, terpenes and flavonoids in cannabis, and potential roles in migraine, headache, and pain: an update on current evidence and cannabis science. Headache In Press Baron EP (2018) Medicinal properties of cannabinoids, terpenes and flavonoids in cannabis, and potential roles in migraine, headache, and pain: an update on current evidence and cannabis science. Headache In Press
29.
go back to reference Ilgen MA, Bohnert K, Kleinberg F et al (2013) Characteristics of adults seeking medical marijuana certification. Drug Alcohol Depend 132:654–659PubMedCrossRef Ilgen MA, Bohnert K, Kleinberg F et al (2013) Characteristics of adults seeking medical marijuana certification. Drug Alcohol Depend 132:654–659PubMedCrossRef
30.
go back to reference Hazekamp A, Heerdink ER (2013) The prevalence and incidence of medicinal cannabis on prescription in the Netherlands. Eur J Clin Pharmacol 69:1575–1580PubMedCrossRef Hazekamp A, Heerdink ER (2013) The prevalence and incidence of medicinal cannabis on prescription in the Netherlands. Eur J Clin Pharmacol 69:1575–1580PubMedCrossRef
41.
go back to reference Grinspoon L, Bakalar JB (1993) Marihuana: the forbidden medicine. Yale University, New Haven Grinspoon L, Bakalar JB (1993) Marihuana: the forbidden medicine. Yale University, New Haven
42.
go back to reference Volfe Z, Dvilansky A, Nathan I (1985) Cannabinoids block release of serotonin from platelets induced by plasma from migraine patients. Int J Clin Pharmacol Res 5:243–246PubMed Volfe Z, Dvilansky A, Nathan I (1985) Cannabinoids block release of serotonin from platelets induced by plasma from migraine patients. Int J Clin Pharmacol Res 5:243–246PubMed
44.
go back to reference Schnelle M, Grotenhermen F, Reif M, Gorter RW (1999) Results of a standardized survey on the medical use of cannabis products in the German-speaking area. Forsch Komplementarmed 6(Suppl 3):28–36PubMed Schnelle M, Grotenhermen F, Reif M, Gorter RW (1999) Results of a standardized survey on the medical use of cannabis products in the German-speaking area. Forsch Komplementarmed 6(Suppl 3):28–36PubMed
45.
go back to reference Gorji A (2003) Pharmacological treatment of headache using traditional Persian medicine. Trends Pharmacol Sci 24:331–334PubMedCrossRef Gorji A (2003) Pharmacological treatment of headache using traditional Persian medicine. Trends Pharmacol Sci 24:331–334PubMedCrossRef
46.
go back to reference Greco R, Gasperi V, Maccarrone M, Tassorelli C (2010) The endocannabinoid system and migraine. Exp Neurol 224:85–91PubMedCrossRef Greco R, Gasperi V, Maccarrone M, Tassorelli C (2010) The endocannabinoid system and migraine. Exp Neurol 224:85–91PubMedCrossRef
47.
go back to reference Rhyne DN, Anderson SL, Gedde M, Borgelt LM (2016) Effects of medical marijuana on migraine headache frequency in an adult population. Pharmacotherapy 36:505–510PubMedCrossRef Rhyne DN, Anderson SL, Gedde M, Borgelt LM (2016) Effects of medical marijuana on migraine headache frequency in an adult population. Pharmacotherapy 36:505–510PubMedCrossRef
48.
go back to reference Donovan M (1845) On the physical and medicinal qualities of Indian hemp (Cannabis Indica); with observations on the best mode of administration, and cases illustrative of its powers. Dublin J Med Sci 26:368–461CrossRef Donovan M (1845) On the physical and medicinal qualities of Indian hemp (Cannabis Indica); with observations on the best mode of administration, and cases illustrative of its powers. Dublin J Med Sci 26:368–461CrossRef
49.
go back to reference Reynolds JR (1868) On some of the therapeutical uses of Indian hemp. Arch Med 2:154–160 Reynolds JR (1868) On some of the therapeutical uses of Indian hemp. Arch Med 2:154–160
50.
go back to reference Waring EJ (1874) Practical therapeutics. Lindsay & Blakiston, Philadelphia Waring EJ (1874) Practical therapeutics. Lindsay & Blakiston, Philadelphia
51.
go back to reference Russo E (2001) Hemp for headache: an in-depth historical and scientific review of cannabis in migraine treatment. J Cannabis Ther 1:21–92CrossRef Russo E (2001) Hemp for headache: an in-depth historical and scientific review of cannabis in migraine treatment. J Cannabis Ther 1:21–92CrossRef
52.
go back to reference Ringer S (1886) A handbook of therapeutics. H.K. Lewis, London Ringer S (1886) A handbook of therapeutics. H.K. Lewis, London
53.
go back to reference Hare HA (1887) Clinical and physiological notes on the action of cannabis Indica. There Gaz 11:225–228 Hare HA (1887) Clinical and physiological notes on the action of cannabis Indica. There Gaz 11:225–228
54.
go back to reference Suckling C (1891) On the therapeutic value of Indian hemp. Br Med J 2:11–12 Suckling C (1891) On the therapeutic value of Indian hemp. Br Med J 2:11–12
55.
go back to reference Mikuriya TH (1991) Chronic migraine headache: five cases successfully treated with marinol and/or illicit cannabis. Schaffer Library of Drug Policy, Berkeley Mikuriya TH (1991) Chronic migraine headache: five cases successfully treated with marinol and/or illicit cannabis. Schaffer Library of Drug Policy, Berkeley
56.
go back to reference Nicolodi M, Sandoval V, Terrine A. Therapeutic use of cannabinoids - dose finding, effects, and pilot data of effects in chronic migraine and cluster headache. Abstract presentation at 3rd congress of the European Academy of Neurology (EAN), Amsterdam, 2017 Nicolodi M, Sandoval V, Terrine A. Therapeutic use of cannabinoids - dose finding, effects, and pilot data of effects in chronic migraine and cluster headache. Abstract presentation at 3rd congress of the European Academy of Neurology (EAN), Amsterdam, 2017
57.
go back to reference Robbins MS, Tarshish S, Solomon S, Grosberg BM (2009) Cluster attacks responsive to recreational cannabis and dronabinol. Headache 49:914–916PubMedCrossRef Robbins MS, Tarshish S, Solomon S, Grosberg BM (2009) Cluster attacks responsive to recreational cannabis and dronabinol. Headache 49:914–916PubMedCrossRef
58.
go back to reference Leroux E, Taifas I, Valade D, Donnet A, Chagnon M, Ducros A (2013) Use of cannabis among 139 cluster headache sufferers. Cephalalgia 33:208–213PubMedCrossRef Leroux E, Taifas I, Valade D, Donnet A, Chagnon M, Ducros A (2013) Use of cannabis among 139 cluster headache sufferers. Cephalalgia 33:208–213PubMedCrossRef
59.
go back to reference Donnet A, Lanteri-Minet M, Guegan-Massardier E et al (2007) Chronic cluster headache: a French clinical descriptive study. J Neurol Neurosurg Psychiatry 78:1354–1358PubMedPubMedCentralCrossRef Donnet A, Lanteri-Minet M, Guegan-Massardier E et al (2007) Chronic cluster headache: a French clinical descriptive study. J Neurol Neurosurg Psychiatry 78:1354–1358PubMedPubMedCentralCrossRef
61.
go back to reference Nunberg H, Kilmer B, Pacula RL, Burgdorf J (2011) An analysis of applicants presenting to a medical marijuana specialty practice in California. J Drug Policy Anal 4(1):1-14 Nunberg H, Kilmer B, Pacula RL, Burgdorf J (2011) An analysis of applicants presenting to a medical marijuana specialty practice in California. J Drug Policy Anal 4(1):1-14
62.
go back to reference Pini LA, Guerzoni S, Cainazzo MM et al (2012) Nabilone for the treatment of medication overuse headache: results of a preliminary double-blind, active-controlled, randomized trial. J Headache Pain 13:677–684PubMedPubMedCentralCrossRef Pini LA, Guerzoni S, Cainazzo MM et al (2012) Nabilone for the treatment of medication overuse headache: results of a preliminary double-blind, active-controlled, randomized trial. J Headache Pain 13:677–684PubMedPubMedCentralCrossRef
63.
go back to reference Evans RW, Ramadan NM (2004) Are cannabis-based chemicals helpful in headache? Headache 44:726–727PubMedCrossRef Evans RW, Ramadan NM (2004) Are cannabis-based chemicals helpful in headache? Headache 44:726–727PubMedCrossRef
64.
go back to reference Consroe P, Musty R, Rein J, Tillery W, Pertwee R (1997) The perceived effects of smoked cannabis on patients with multiple sclerosis. Eur Neurol 38:44–48PubMedCrossRef Consroe P, Musty R, Rein J, Tillery W, Pertwee R (1997) The perceived effects of smoked cannabis on patients with multiple sclerosis. Eur Neurol 38:44–48PubMedCrossRef
67.
go back to reference Lipton RB, Dodick D, Sadovsky R et al (2003) A self-administered screener for migraine in primary care: the ID migraine validation study. Neurology 61:375–382PubMedCrossRef Lipton RB, Dodick D, Sadovsky R et al (2003) A self-administered screener for migraine in primary care: the ID migraine validation study. Neurology 61:375–382PubMedCrossRef
68.
go back to reference Akerman S, Holland PR, Lasalandra MP, Goadsby PJ (2013) Endocannabinoids in the brainstem modulate dural trigeminovascular nociceptive traffic via CB1 and “triptan” receptors: implications in migraine. J Neurosci 33:14869–14877PubMedPubMedCentralCrossRef Akerman S, Holland PR, Lasalandra MP, Goadsby PJ (2013) Endocannabinoids in the brainstem modulate dural trigeminovascular nociceptive traffic via CB1 and “triptan” receptors: implications in migraine. J Neurosci 33:14869–14877PubMedPubMedCentralCrossRef
69.
go back to reference Akerman S, Holland PR, Goadsby PJ (2007) Cannabinoid (CB1) receptor activation inhibits trigeminovascular neurons. J Pharmacol Exp Ther 320:64–71PubMedCrossRef Akerman S, Holland PR, Goadsby PJ (2007) Cannabinoid (CB1) receptor activation inhibits trigeminovascular neurons. J Pharmacol Exp Ther 320:64–71PubMedCrossRef
70.
go back to reference Akerman S, Kaube H, Goadsby PJ (2004) Anandamide is able to inhibit trigeminal neurons using an in vivo model of trigeminovascular-mediated nociception. J Pharmacol Exp Ther 309:56–63PubMedCrossRef Akerman S, Kaube H, Goadsby PJ (2004) Anandamide is able to inhibit trigeminal neurons using an in vivo model of trigeminovascular-mediated nociception. J Pharmacol Exp Ther 309:56–63PubMedCrossRef
71.
72.
go back to reference Rodriguez de Fonseca F, Del Arco I, Bermudez-Silva FJ, Bilbao A, Cippitelli A, Navarro M (2005) The endocannabinoid system: physiology and pharmacology. Alcohol Alcohol 40:2–14PubMedCrossRef Rodriguez de Fonseca F, Del Arco I, Bermudez-Silva FJ, Bilbao A, Cippitelli A, Navarro M (2005) The endocannabinoid system: physiology and pharmacology. Alcohol Alcohol 40:2–14PubMedCrossRef
73.
go back to reference Maccarrone M, Gasperi V, Catani MV et al (2010) The endocannabinoid system and its relevance for nutrition. Annu Rev Nutr 30:423–440PubMedCrossRef Maccarrone M, Gasperi V, Catani MV et al (2010) The endocannabinoid system and its relevance for nutrition. Annu Rev Nutr 30:423–440PubMedCrossRef
75.
go back to reference Mallat A, Teixeira-Clerc F, Deveaux V, Manin S, Lotersztajn S (2011) The endocannabinoid system as a key mediator during liver diseases: new insights and therapeutic openings. Br J Pharmacol 163:1432–1440PubMedPubMedCentralCrossRef Mallat A, Teixeira-Clerc F, Deveaux V, Manin S, Lotersztajn S (2011) The endocannabinoid system as a key mediator during liver diseases: new insights and therapeutic openings. Br J Pharmacol 163:1432–1440PubMedPubMedCentralCrossRef
76.
go back to reference Galve-Roperh I, Rueda D, Gomez del Pulgar T, Velasco G, Guzman M (2002) Mechanism of extracellular signal-regulated kinase activation by the CB(1) cannabinoid receptor. Mol Pharmacol 62:1385–1392PubMedCrossRef Galve-Roperh I, Rueda D, Gomez del Pulgar T, Velasco G, Guzman M (2002) Mechanism of extracellular signal-regulated kinase activation by the CB(1) cannabinoid receptor. Mol Pharmacol 62:1385–1392PubMedCrossRef
78.
go back to reference De Petrocellis L, Di Marzo V (2009) An introduction to the endocannabinoid system: from the early to the latest concepts. Best Pract Res Clin Endocrinol Metab 23:1–15PubMedCrossRef De Petrocellis L, Di Marzo V (2009) An introduction to the endocannabinoid system: from the early to the latest concepts. Best Pract Res Clin Endocrinol Metab 23:1–15PubMedCrossRef
79.
go back to reference Devane WA, Hanus L, Breuer A et al (1992) Isolation and structure of a brain constituent that binds to the cannabinoid receptor. Science 258:1946–1949PubMedCrossRef Devane WA, Hanus L, Breuer A et al (1992) Isolation and structure of a brain constituent that binds to the cannabinoid receptor. Science 258:1946–1949PubMedCrossRef
82.
go back to reference Di Marzo V, Petrocellis LD (2006) Plant, synthetic, and endogenous cannabinoids in medicine. Annu Rev Med 57:553–574PubMedCrossRef Di Marzo V, Petrocellis LD (2006) Plant, synthetic, and endogenous cannabinoids in medicine. Annu Rev Med 57:553–574PubMedCrossRef
83.
go back to reference Pertwee RG (2008) The diverse CB1 and CB2 receptor pharmacology of three plant cannabinoids: delta9-tetrahydrocannabinol, cannabidiol and delta9-tetrahydrocannabivarin. Br J Pharmacol 153:199–215PubMedCrossRef Pertwee RG (2008) The diverse CB1 and CB2 receptor pharmacology of three plant cannabinoids: delta9-tetrahydrocannabinol, cannabidiol and delta9-tetrahydrocannabivarin. Br J Pharmacol 153:199–215PubMedCrossRef
85.
go back to reference Guindon J, Beaulieu P (2009) The role of the endogenous cannabinoid system in peripheral analgesia. Curr Mol Pharmacol 2:134–139PubMedCrossRef Guindon J, Beaulieu P (2009) The role of the endogenous cannabinoid system in peripheral analgesia. Curr Mol Pharmacol 2:134–139PubMedCrossRef
86.
go back to reference Kraft B (2012) Is there any clinically relevant cannabinoid-induced analgesia? Pharmacology 89:237–246PubMedCrossRef Kraft B (2012) Is there any clinically relevant cannabinoid-induced analgesia? Pharmacology 89:237–246PubMedCrossRef
87.
go back to reference Ramikie TS, Nyilas R, Bluett RJ et al (2014) Multiple mechanistically distinct modes of endocannabinoid mobilization at central amygdala glutamatergic synapses. Neuron 81:1111–1125PubMedPubMedCentralCrossRef Ramikie TS, Nyilas R, Bluett RJ et al (2014) Multiple mechanistically distinct modes of endocannabinoid mobilization at central amygdala glutamatergic synapses. Neuron 81:1111–1125PubMedPubMedCentralCrossRef
89.
go back to reference Pertwee RG, Howlett AC, Abood ME et al (2010) International Union of Basic and Clinical Pharmacology. LXXIX. Cannabinoid receptors and their ligands: beyond CB(1) and CB(2). Pharmacol Rev 62:588–631PubMedPubMedCentralCrossRef Pertwee RG, Howlett AC, Abood ME et al (2010) International Union of Basic and Clinical Pharmacology. LXXIX. Cannabinoid receptors and their ligands: beyond CB(1) and CB(2). Pharmacol Rev 62:588–631PubMedPubMedCentralCrossRef
90.
go back to reference Katona I, Freund TF (2008) Endocannabinoid signaling as a synaptic circuit breaker in neurological disease. Nat Med 14:923–930PubMedCrossRef Katona I, Freund TF (2008) Endocannabinoid signaling as a synaptic circuit breaker in neurological disease. Nat Med 14:923–930PubMedCrossRef
91.
go back to reference Raichlen DA, Foster AD, Gerdeman GL, Seillier A, Giuffrida A (2012) Wired to run: exercise-induced endocannabinoid signaling in humans and cursorial mammals with implications for the ‘runner’s high’. J Exp Biol 215:1331–1336PubMedCrossRef Raichlen DA, Foster AD, Gerdeman GL, Seillier A, Giuffrida A (2012) Wired to run: exercise-induced endocannabinoid signaling in humans and cursorial mammals with implications for the ‘runner’s high’. J Exp Biol 215:1331–1336PubMedCrossRef
93.
go back to reference Napchan U, Buse DC, Loder EW (2011) The use of marijuana or synthetic cannabinoids for the treatment of headache. Headache 51:502–505PubMedCrossRef Napchan U, Buse DC, Loder EW (2011) The use of marijuana or synthetic cannabinoids for the treatment of headache. Headache 51:502–505PubMedCrossRef
95.
go back to reference Klein TW, Cabral GA (2006) Cannabinoid-induced immune suppression and modulation of antigen-presenting cells. J NeuroImmune Pharmacol 1:50–64PubMedCrossRef Klein TW, Cabral GA (2006) Cannabinoid-induced immune suppression and modulation of antigen-presenting cells. J NeuroImmune Pharmacol 1:50–64PubMedCrossRef
96.
go back to reference Zhang HY, Gao M, Liu QR et al (2014) Cannabinoid CB2 receptors modulate midbrain dopamine neuronal activity and dopamine-related behavior in mice. Proc Natl Acad Sci U S A 111:E5007–E5015PubMedPubMedCentralCrossRef Zhang HY, Gao M, Liu QR et al (2014) Cannabinoid CB2 receptors modulate midbrain dopamine neuronal activity and dopamine-related behavior in mice. Proc Natl Acad Sci U S A 111:E5007–E5015PubMedPubMedCentralCrossRef
97.
go back to reference Zhang HY, Gao M, Shen H et al (2017) Expression of functional cannabinoid CB2 receptor in VTA dopamine neurons in rats. Addict Biol 22:752–765PubMedCrossRef Zhang HY, Gao M, Shen H et al (2017) Expression of functional cannabinoid CB2 receptor in VTA dopamine neurons in rats. Addict Biol 22:752–765PubMedCrossRef
101.
102.
go back to reference Hajos N, Ledent C, Freund TF (2001) Novel cannabinoid-sensitive receptor mediates inhibition of glutamatergic synaptic transmission in the hippocampus. Neuroscience 106:1–4PubMedCrossRef Hajos N, Ledent C, Freund TF (2001) Novel cannabinoid-sensitive receptor mediates inhibition of glutamatergic synaptic transmission in the hippocampus. Neuroscience 106:1–4PubMedCrossRef
103.
go back to reference Govaerts SJ, Hermans E, Lambert DM (2004) Comparison of cannabinoid ligands affinities and efficacies in murine tissues and in transfected cells expressing human recombinant cannabinoid receptors. Eur J Pharm Sci 23:233–243PubMedCrossRef Govaerts SJ, Hermans E, Lambert DM (2004) Comparison of cannabinoid ligands affinities and efficacies in murine tissues and in transfected cells expressing human recombinant cannabinoid receptors. Eur J Pharm Sci 23:233–243PubMedCrossRef
104.
go back to reference Marcu JP (2016) An overview of major and minor phytocannabinoids. In: Preedy V (ed) Neuropathology of drug addictions and substance misuse, Volume 1: foundations of understanding, tobacco, alcohol, cannabinoids and opioids. Academic Press, London, pp 672–678CrossRef Marcu JP (2016) An overview of major and minor phytocannabinoids. In: Preedy V (ed) Neuropathology of drug addictions and substance misuse, Volume 1: foundations of understanding, tobacco, alcohol, cannabinoids and opioids. Academic Press, London, pp 672–678CrossRef
105.
go back to reference O'Sullivan SE, Kendall DA, Randall MD (2009) Time-dependent vascular effects of endocannabinoids mediated by peroxisome proliferator-activated receptor gamma (PPARgamma). PPAR Res 2009:425289PubMedPubMedCentralCrossRef O'Sullivan SE, Kendall DA, Randall MD (2009) Time-dependent vascular effects of endocannabinoids mediated by peroxisome proliferator-activated receptor gamma (PPARgamma). PPAR Res 2009:425289PubMedPubMedCentralCrossRef
106.
go back to reference O'Sullivan SE, Kendall DA (2010) Cannabinoid activation of peroxisome proliferator-activated receptors: potential for modulation of inflammatory disease. Immunobiology 215:611–616PubMedCrossRef O'Sullivan SE, Kendall DA (2010) Cannabinoid activation of peroxisome proliferator-activated receptors: potential for modulation of inflammatory disease. Immunobiology 215:611–616PubMedCrossRef
107.
go back to reference De Petrocellis L, Orlando P, Moriello AS et al (2012) Cannabinoid actions at TRPV channels: effects on TRPV3 and TRPV4 and their potential relevance to gastrointestinal inflammation. Acta Physiol (Oxf) 204:255–266CrossRef De Petrocellis L, Orlando P, Moriello AS et al (2012) Cannabinoid actions at TRPV channels: effects on TRPV3 and TRPV4 and their potential relevance to gastrointestinal inflammation. Acta Physiol (Oxf) 204:255–266CrossRef
108.
go back to reference De Petrocellis L, Vellani V, Schiano-Moriello A et al (2008) Plant-derived cannabinoids modulate the activity of transient receptor potential channels of ankyrin type-1 and melastatin type-8. J Pharmacol Exp Ther 325:1007–1015PubMedCrossRef De Petrocellis L, Vellani V, Schiano-Moriello A et al (2008) Plant-derived cannabinoids modulate the activity of transient receptor potential channels of ankyrin type-1 and melastatin type-8. J Pharmacol Exp Ther 325:1007–1015PubMedCrossRef
109.
go back to reference De Petrocellis L, Di Marzo V (2010) Non-CB1, non-CB2 receptors for endocannabinoids, plant cannabinoids, and synthetic cannabimimetics: focus on G-protein-coupled receptors and transient receptor potential channels. J NeuroImmune Pharmacol 5:103–121PubMedCrossRef De Petrocellis L, Di Marzo V (2010) Non-CB1, non-CB2 receptors for endocannabinoids, plant cannabinoids, and synthetic cannabimimetics: focus on G-protein-coupled receptors and transient receptor potential channels. J NeuroImmune Pharmacol 5:103–121PubMedCrossRef
110.
go back to reference De Petrocellis L, Ligresti A, Moriello AS et al (2011) Effects of cannabinoids and cannabinoid-enriched Cannabis extracts on TRP channels and endocannabinoid metabolic enzymes. Br J Pharmacol 163:1479–1494PubMedPubMedCentralCrossRef De Petrocellis L, Ligresti A, Moriello AS et al (2011) Effects of cannabinoids and cannabinoid-enriched Cannabis extracts on TRP channels and endocannabinoid metabolic enzymes. Br J Pharmacol 163:1479–1494PubMedPubMedCentralCrossRef
111.
go back to reference Barann M, Molderings G, Bruss M, Bonisch H, Urban BW, Gothert M (2002) Direct inhibition by cannabinoids of human 5-HT3A receptors: probable involvement of an allosteric modulatory site. Br J Pharmacol 137:589–596PubMedPubMedCentralCrossRef Barann M, Molderings G, Bruss M, Bonisch H, Urban BW, Gothert M (2002) Direct inhibition by cannabinoids of human 5-HT3A receptors: probable involvement of an allosteric modulatory site. Br J Pharmacol 137:589–596PubMedPubMedCentralCrossRef
112.
go back to reference Hejazi N, Zhou C, Oz M, Sun H, Ye JH, Zhang L (2006) Delta9-tetrahydrocannabinol and endogenous cannabinoid anandamide directly potentiate the function of glycine receptors. Mol Pharmacol 69:991–997PubMed Hejazi N, Zhou C, Oz M, Sun H, Ye JH, Zhang L (2006) Delta9-tetrahydrocannabinol and endogenous cannabinoid anandamide directly potentiate the function of glycine receptors. Mol Pharmacol 69:991–997PubMed
113.
go back to reference McHugh D, Page J, Dunn E, Bradshaw HB (2012) Delta(9) -tetrahydrocannabinol and N-arachidonyl glycine are full agonists at GPR18 receptors and induce migration in human endometrial HEC-1B cells. Br J Pharmacol 165:2414–2424PubMedPubMedCentralCrossRef McHugh D, Page J, Dunn E, Bradshaw HB (2012) Delta(9) -tetrahydrocannabinol and N-arachidonyl glycine are full agonists at GPR18 receptors and induce migration in human endometrial HEC-1B cells. Br J Pharmacol 165:2414–2424PubMedPubMedCentralCrossRef
114.
go back to reference Hampson AJ, Grimaldi M, Axelrod J, Wink D (1998) Cannabidiol and (−)Delta9-tetrahydrocannabinol are neuroprotective antioxidants. Proc Natl Acad Sci U S A 95:8268–8273PubMedPubMedCentralCrossRef Hampson AJ, Grimaldi M, Axelrod J, Wink D (1998) Cannabidiol and (−)Delta9-tetrahydrocannabinol are neuroprotective antioxidants. Proc Natl Acad Sci U S A 95:8268–8273PubMedPubMedCentralCrossRef
115.
go back to reference Hampson AJ, Bornheim LM, Scanziani M et al (1998) Dual effects of anandamide on NMDA receptor-mediated responses and neurotransmission. J Neurochem 70:671–676PubMedCrossRef Hampson AJ, Bornheim LM, Scanziani M et al (1998) Dual effects of anandamide on NMDA receptor-mediated responses and neurotransmission. J Neurochem 70:671–676PubMedCrossRef
116.
go back to reference Hampson AJ, Grimaldi M, Lolic M, Wink D, Rosenthal R, Axelrod J (2000) Neuroprotective antioxidants from marijuana. Ann N Y Acad Sci 899:274–282PubMedCrossRef Hampson AJ, Grimaldi M, Lolic M, Wink D, Rosenthal R, Axelrod J (2000) Neuroprotective antioxidants from marijuana. Ann N Y Acad Sci 899:274–282PubMedCrossRef
117.
go back to reference Li J, Daughters RS, Bullis C et al (1999) The cannabinoid receptor agonist WIN 55,212-2 mesylate blocks the development of hyperalgesia produced by capsaicin in rats. Pain 81:25–33PubMedCrossRef Li J, Daughters RS, Bullis C et al (1999) The cannabinoid receptor agonist WIN 55,212-2 mesylate blocks the development of hyperalgesia produced by capsaicin in rats. Pain 81:25–33PubMedCrossRef
118.
121.
go back to reference Welch SP (1993) 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 265:633–640PubMed Welch SP (1993) 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 265:633–640PubMed
122.
go back to reference Smith FL, Cichewicz D, Martin ZL, Welch SP (1998) The enhancement of morphine antinociception in mice by delta9-tetrahydrocannabinol. Pharmacol Biochem Behav 60:559–566PubMedCrossRef Smith FL, Cichewicz D, Martin ZL, Welch SP (1998) The enhancement of morphine antinociception in mice by delta9-tetrahydrocannabinol. Pharmacol Biochem Behav 60:559–566PubMedCrossRef
123.
go back to reference Smith PA, Selley DE, Sim-Selley LJ, Welch SP (2007) Low dose combination of morphine and delta9-tetrahydrocannabinol circumvents antinociceptive tolerance and apparent desensitization of receptors. Eur J Pharmacol 571:129–137PubMedPubMedCentralCrossRef Smith PA, Selley DE, Sim-Selley LJ, Welch SP (2007) Low dose combination of morphine and delta9-tetrahydrocannabinol circumvents antinociceptive tolerance and apparent desensitization of receptors. Eur J Pharmacol 571:129–137PubMedPubMedCentralCrossRef
124.
go back to reference Cichewicz DL, McCarthy EA (2003) Antinociceptive synergy between delta(9)-tetrahydrocannabinol and opioids after oral administration. J Pharmacol Exp Ther 304:1010–1015PubMedCrossRef Cichewicz DL, McCarthy EA (2003) Antinociceptive synergy between delta(9)-tetrahydrocannabinol and opioids after oral administration. J Pharmacol Exp Ther 304:1010–1015PubMedCrossRef
125.
go back to reference Cichewicz DL (2004) Synergistic interactions between cannabinoid and opioid analgesics. Life Sci 74:1317–1324PubMedCrossRef Cichewicz DL (2004) Synergistic interactions between cannabinoid and opioid analgesics. Life Sci 74:1317–1324PubMedCrossRef
126.
go back to reference Manzanares J, Corchero J, Romero J, Fernandez-Ruiz JJ, Ramos JA, Fuentes JA (1998) Chronic administration of cannabinoids regulates proenkephalin mRNA levels in selected regions of the rat brain. Brain Res Mol Brain Res 55:126–132PubMedCrossRef Manzanares J, Corchero J, Romero J, Fernandez-Ruiz JJ, Ramos JA, Fuentes JA (1998) Chronic administration of cannabinoids regulates proenkephalin mRNA levels in selected regions of the rat brain. Brain Res Mol Brain Res 55:126–132PubMedCrossRef
127.
go back to reference Manzanares J, Julian M, Carrascosa A (2006) Role of the cannabinoid system in pain control and therapeutic implications for the management of acute and chronic pain episodes. Curr Neuropharmacol 4:239–257PubMedPubMedCentralCrossRef Manzanares J, Julian M, Carrascosa A (2006) Role of the cannabinoid system in pain control and therapeutic implications for the management of acute and chronic pain episodes. Curr Neuropharmacol 4:239–257PubMedPubMedCentralCrossRef
128.
go back to reference Evans FJ (1991) Cannabinoids: the separation of central from peripheral effects on a structural basis. Planta Med 57:S60–S67CrossRefPubMed Evans FJ (1991) Cannabinoids: the separation of central from peripheral effects on a structural basis. Planta Med 57:S60–S67CrossRefPubMed
130.
go back to reference Izzo AA, Borrelli F, Capasso R, Di Marzo V, Mechoulam R (2009) Non-psychotropic plant cannabinoids: new therapeutic opportunities from an ancient herb. Trends Pharmacol Sci 30:515–527PubMedCrossRef Izzo AA, Borrelli F, Capasso R, Di Marzo V, Mechoulam R (2009) Non-psychotropic plant cannabinoids: new therapeutic opportunities from an ancient herb. Trends Pharmacol Sci 30:515–527PubMedCrossRef
131.
133.
go back to reference Noyes R Jr, Brunk SF, Avery DA, Canter AC (1975) The analgesic properties of delta-9-tetrahydrocannabinol and codeine. Clin Pharmacol Ther 18:84–89PubMedCrossRef Noyes R Jr, Brunk SF, Avery DA, Canter AC (1975) The analgesic properties of delta-9-tetrahydrocannabinol and codeine. Clin Pharmacol Ther 18:84–89PubMedCrossRef
134.
go back to reference Noyes R Jr, Brunk SF, Baram DA, Canter A (1975) Analgesic effect of delta-9-tetrahydrocannabinol. J Clin Pharmacol 15:139–143PubMedCrossRef Noyes R Jr, Brunk SF, Baram DA, Canter A (1975) Analgesic effect of delta-9-tetrahydrocannabinol. J Clin Pharmacol 15:139–143PubMedCrossRef
135.
go back to reference Schley M, Legler A, Skopp G, Schmelz M, Konrad C, Rukwied R (2006) Delta-9-THC based monotherapy in fibromyalgia patients on experimentally induced pain, axon reflex flare, and pain relief. Curr Med Res Opin 22:1269–1276PubMedCrossRef Schley M, Legler A, Skopp G, Schmelz M, Konrad C, Rukwied R (2006) Delta-9-THC based monotherapy in fibromyalgia patients on experimentally induced pain, axon reflex flare, and pain relief. Curr Med Res Opin 22:1269–1276PubMedCrossRef
136.
go back to reference Maurer M, Henn V, Dittrich A, Hofmann A (1990) Delta-9-tetrahydrocannabinol shows antispastic and analgesic effects in a single case double-blind trial. Eur Arch Psychiatry Clin Neurosci 240:1–4PubMedCrossRef Maurer M, Henn V, Dittrich A, Hofmann A (1990) Delta-9-tetrahydrocannabinol shows antispastic and analgesic effects in a single case double-blind trial. Eur Arch Psychiatry Clin Neurosci 240:1–4PubMedCrossRef
137.
go back to reference Srivastava MD, Srivastava BI, Brouhard B (1998) Delta9 tetrahydrocannabinol and cannabidiol alter cytokine production by human immune cells. Immunopharmacology 40:179–185PubMedCrossRef Srivastava MD, Srivastava BI, Brouhard B (1998) Delta9 tetrahydrocannabinol and cannabidiol alter cytokine production by human immune cells. Immunopharmacology 40:179–185PubMedCrossRef
138.
go back to reference Kozela E, Pietr M, Juknat A, Rimmerman N, Levy R, Vogel Z (2010) 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 285:1616–1626PubMedCrossRef Kozela E, Pietr M, Juknat A, Rimmerman N, Levy R, Vogel Z (2010) 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 285:1616–1626PubMedCrossRef
139.
140.
go back to reference Ruhaak LR, Felth J, Karlsson PC, Rafter JJ, Verpoorte R, Bohlin L (2011) Evaluation of the cyclooxygenase inhibiting effects of six major cannabinoids isolated from cannabis sativa. Biol Pharm Bull 34:774–778PubMedCrossRef Ruhaak LR, Felth J, Karlsson PC, Rafter JJ, Verpoorte R, Bohlin L (2011) Evaluation of the cyclooxygenase inhibiting effects of six major cannabinoids isolated from cannabis sativa. Biol Pharm Bull 34:774–778PubMedCrossRef
141.
go back to reference Martin BR, Compton DR, Thomas BF et al (1991) Behavioral, biochemical, and molecular modeling evaluations of cannabinoid analogs. Pharmacol Biochem Behav 40:471–478PubMedCrossRef Martin BR, Compton DR, Thomas BF et al (1991) Behavioral, biochemical, and molecular modeling evaluations of cannabinoid analogs. Pharmacol Biochem Behav 40:471–478PubMedCrossRef
142.
go back to reference Martin BR, Lichtman AH (1998) Cannabinoid transmission and pain perception. Neurobiol Dis 5:447–461PubMedCrossRef Martin BR, Lichtman AH (1998) Cannabinoid transmission and pain perception. Neurobiol Dis 5:447–461PubMedCrossRef
143.
go back to reference DeLong GT, Wolf CE, Poklis A, Lichtman AH (2010) Pharmacological evaluation of the natural constituent of cannabis sativa, cannabichromene and its modulation by Delta(9)-tetrahydrocannabinol. Drug Alcohol Depend 112:126–133PubMedPubMedCentralCrossRef DeLong GT, Wolf CE, Poklis A, Lichtman AH (2010) Pharmacological evaluation of the natural constituent of cannabis sativa, cannabichromene and its modulation by Delta(9)-tetrahydrocannabinol. Drug Alcohol Depend 112:126–133PubMedPubMedCentralCrossRef
144.
go back to reference Zygmunt PM, Andersson DA, Hogestatt ED (2002) Delta 9-tetrahydrocannabinol and cannabinol activate capsaicin-sensitive sensory nerves via a CB1 and CB2 cannabinoid receptor-independent mechanism. J Neurosci 22:4720–4727PubMedCrossRef Zygmunt PM, Andersson DA, Hogestatt ED (2002) Delta 9-tetrahydrocannabinol and cannabinol activate capsaicin-sensitive sensory nerves via a CB1 and CB2 cannabinoid receptor-independent mechanism. J Neurosci 22:4720–4727PubMedCrossRef
145.
go back to reference Burston JJ, Sagar DR, Shao P et al (2013) Cannabinoid CB2 receptors regulate central sensitization and pain responses associated with osteoarthritis of the knee joint. PLoS One 8:e80440PubMedPubMedCentralCrossRef Burston JJ, Sagar DR, Shao P et al (2013) Cannabinoid CB2 receptors regulate central sensitization and pain responses associated with osteoarthritis of the knee joint. PLoS One 8:e80440PubMedPubMedCentralCrossRef
146.
go back to reference Gui H, Liu X, Wang ZW, He DY, Su DF, Dai SM (2014) Expression of cannabinoid receptor 2 and its inhibitory effects on synovial fibroblasts in rheumatoid arthritis. Rheumatology (Oxford) 53:802–809CrossRef Gui H, Liu X, Wang ZW, He DY, Su DF, Dai SM (2014) Expression of cannabinoid receptor 2 and its inhibitory effects on synovial fibroblasts in rheumatoid arthritis. Rheumatology (Oxford) 53:802–809CrossRef
147.
go back to reference Clayton N, Marshall FH, Bountra C, O'Shaughnessy CT (2002) CB1 and CB2 cannabinoid receptors are implicated in inflammatory pain. Pain 96:253–260PubMedCrossRef Clayton N, Marshall FH, Bountra C, O'Shaughnessy CT (2002) CB1 and CB2 cannabinoid receptors are implicated in inflammatory pain. Pain 96:253–260PubMedCrossRef
148.
149.
go back to reference Jensen B, Chen J, Furnish T, Wallace M (2015) Medical marijuana and chronic pain: a review of basic science and clinical evidence. Curr Pain Headache Rep 19:50PubMedCrossRef Jensen B, Chen J, Furnish T, Wallace M (2015) Medical marijuana and chronic pain: a review of basic science and clinical evidence. Curr Pain Headache Rep 19:50PubMedCrossRef
150.
go back to reference Weber J, Schley M, Casutt M et al (2009) Tetrahydrocannabinol (Delta 9-THC) treatment in chronic central neuropathic pain and fibromyalgia patients: results of a multicenter survey. Anesthesiol Res Pract 2009. https://doi.org/10.1155/2009/827290 Epub 2009 Oct 25 Weber J, Schley M, Casutt M et al (2009) Tetrahydrocannabinol (Delta 9-THC) treatment in chronic central neuropathic pain and fibromyalgia patients: results of a multicenter survey. Anesthesiol Res Pract 2009. https://​doi.​org/​10.​1155/​2009/​827290 Epub 2009 Oct 25
151.
go back to reference Weber M, Goldman B, Truniger S (2010) Tetrahydrocannabinol (THC) for cramps in amyotrophic lateral sclerosis: a randomised, double-blind crossover trial. J Neurol Neurosurg Psychiatry 81:1135–1140PubMedCrossRef Weber M, Goldman B, Truniger S (2010) Tetrahydrocannabinol (THC) for cramps in amyotrophic lateral sclerosis: a randomised, double-blind crossover trial. J Neurol Neurosurg Psychiatry 81:1135–1140PubMedCrossRef
152.
go back to reference Smith PB, Martin BR (1992) Spinal mechanisms of delta 9-tetrahydrocannabinol-induced analgesia. Brain Res 578:8–12PubMedCrossRef Smith PB, Martin BR (1992) Spinal mechanisms of delta 9-tetrahydrocannabinol-induced analgesia. Brain Res 578:8–12PubMedCrossRef
153.
go back to reference Smith FL, Fujimori K, Lowe J, Welch SP (1998) Characterization of delta9-tetrahydrocannabinol and anandamide antinociception in nonarthritic and arthritic rats. Pharmacol Biochem Behav 60:183–191PubMedCrossRef Smith FL, Fujimori K, Lowe J, Welch SP (1998) Characterization of delta9-tetrahydrocannabinol and anandamide antinociception in nonarthritic and arthritic rats. Pharmacol Biochem Behav 60:183–191PubMedCrossRef
155.
156.
go back to reference Walker JM, Hohmann AG, Martin WJ, Strangman NM, Huang SM, Tsou K (1999) The neurobiology of cannabinoid analgesia. Life Sci 65:665–673PubMedCrossRef Walker JM, Hohmann AG, Martin WJ, Strangman NM, Huang SM, Tsou K (1999) The neurobiology of cannabinoid analgesia. Life Sci 65:665–673PubMedCrossRef
157.
158.
go back to reference Wilsey B, Marcotte T, Deutsch R, Gouaux B, Sakai S, Donaghe H (2013) Low-dose vaporized cannabis significantly improves neuropathic pain. J Pain 14:136–148PubMedCrossRef Wilsey B, Marcotte T, Deutsch R, Gouaux B, Sakai S, Donaghe H (2013) Low-dose vaporized cannabis significantly improves neuropathic pain. J Pain 14:136–148PubMedCrossRef
159.
go back to reference Wilsey B, Marcotte T, Tsodikov A et al (2008) A randomized, placebo-controlled, crossover trial of cannabis cigarettes in neuropathic pain. J Pain 9:506–521PubMedPubMedCentralCrossRef Wilsey B, Marcotte T, Tsodikov A et al (2008) A randomized, placebo-controlled, crossover trial of cannabis cigarettes in neuropathic pain. J Pain 9:506–521PubMedPubMedCentralCrossRef
160.
go back to reference Ellis RJ, Toperoff W, Vaida F et al (2009) Smoked medicinal cannabis for neuropathic pain in HIV: a randomized, crossover clinical trial. Neuropsychopharmacology 34:672–680PubMedCrossRef Ellis RJ, Toperoff W, Vaida F et al (2009) Smoked medicinal cannabis for neuropathic pain in HIV: a randomized, crossover clinical trial. Neuropsychopharmacology 34:672–680PubMedCrossRef
161.
go back to reference Abrams DI, Jay CA, Shade SB et al (2007) Cannabis in painful HIV-associated sensory neuropathy: a randomized placebo-controlled trial. Neurology 68:515–521PubMedCrossRef Abrams DI, Jay CA, Shade SB et al (2007) Cannabis in painful HIV-associated sensory neuropathy: a randomized placebo-controlled trial. Neurology 68:515–521PubMedCrossRef
162.
go back to reference Corey-Bloom J, Wolfson T, Gamst A et al (2012) Smoked cannabis for spasticity in multiple sclerosis: a randomized, placebo-controlled trial. CMAJ 184:1143–1150PubMedPubMedCentralCrossRef Corey-Bloom J, Wolfson T, Gamst A et al (2012) Smoked cannabis for spasticity in multiple sclerosis: a randomized, placebo-controlled trial. CMAJ 184:1143–1150PubMedPubMedCentralCrossRef
163.
164.
go back to reference Wallace M, Schulteis G, Atkinson JH et al (2007) Dose-dependent effects of smoked cannabis on capsaicin-induced pain and hyperalgesia in healthy volunteers. Anesthesiology 107:785–796PubMedCrossRef Wallace M, Schulteis G, Atkinson JH et al (2007) Dose-dependent effects of smoked cannabis on capsaicin-induced pain and hyperalgesia in healthy volunteers. Anesthesiology 107:785–796PubMedCrossRef
165.
go back to reference Greenwald MK, Stitzer ML (2000) Antinociceptive, subjective and behavioral effects of smoked marijuana in humans. Drug Alcohol Depend 59:261–275PubMedCrossRef Greenwald MK, Stitzer ML (2000) Antinociceptive, subjective and behavioral effects of smoked marijuana in humans. Drug Alcohol Depend 59:261–275PubMedCrossRef
166.
go back to reference Abrams DI, Couey P, Shade SB, Kelly ME, Benowitz NL (2011) Cannabinoid-opioid interaction in chronic pain. Clin Pharmacol Ther 90:844–851PubMedCrossRef Abrams DI, Couey P, Shade SB, Kelly ME, Benowitz NL (2011) Cannabinoid-opioid interaction in chronic pain. Clin Pharmacol Ther 90:844–851PubMedCrossRef
167.
go back to reference Eisenberg E, Ogintz M, Almog S (2014) The pharmacokinetics, efficacy, safety, and ease of use of a novel portable metered-dose cannabis inhaler in patients with chronic neuropathic pain: a phase 1a study. J Pain Palliat Care Pharmacother 28:216–225PubMedCrossRef Eisenberg E, Ogintz M, Almog S (2014) The pharmacokinetics, efficacy, safety, and ease of use of a novel portable metered-dose cannabis inhaler in patients with chronic neuropathic pain: a phase 1a study. J Pain Palliat Care Pharmacother 28:216–225PubMedCrossRef
168.
go back to reference Ware MA, Wang T, Shapiro S, Collet JP (2015) COMPASS study team. Cannabis for the Management of Pain: Assessment of Safety Study (COMPASS). J Pain 16:1233–1242PubMedCrossRef Ware MA, Wang T, Shapiro S, Collet JP (2015) COMPASS study team. Cannabis for the Management of Pain: Assessment of Safety Study (COMPASS). J Pain 16:1233–1242PubMedCrossRef
169.
go back to reference Fiz J, Duran M, Capella D, Carbonell J, Farre M (2011) Cannabis use in patients with fibromyalgia: effect on symptoms relief and health-related quality of life. PLoS One 6:e18440PubMedPubMedCentralCrossRef Fiz J, Duran M, Capella D, Carbonell J, Farre M (2011) Cannabis use in patients with fibromyalgia: effect on symptoms relief and health-related quality of life. PLoS One 6:e18440PubMedPubMedCentralCrossRef
170.
go back to reference Ben-Shabat S, Fride E, Sheskin T et al (1998) An entourage effect: inactive endogenous fatty acid glycerol esters enhance 2-arachidonoyl-glycerol cannabinoid activity. Eur J Pharmacol 353:23–31PubMedCrossRef Ben-Shabat S, Fride E, Sheskin T et al (1998) An entourage effect: inactive endogenous fatty acid glycerol esters enhance 2-arachidonoyl-glycerol cannabinoid activity. Eur J Pharmacol 353:23–31PubMedCrossRef
171.
go back to reference Nurmikko TJ, Serpell MG, Hoggart B, Toomey PJ, Morlion BJ, Haines D (2007) Sativex successfully treats neuropathic pain characterised by allodynia: a randomised, double-blind, placebo-controlled clinical trial. Pain 133:210–220PubMedCrossRef Nurmikko TJ, Serpell MG, Hoggart B, Toomey PJ, Morlion BJ, Haines D (2007) Sativex successfully treats neuropathic pain characterised by allodynia: a randomised, double-blind, placebo-controlled clinical trial. Pain 133:210–220PubMedCrossRef
172.
go back to reference Rog DJ, Nurmikko TJ, Young CA (2007) Oromucosal delta9-tetrahydrocannabinol/cannabidiol for neuropathic pain associated with multiple sclerosis: an uncontrolled, open-label, 2-year extension trial. Clin Ther 29:2068–2079PubMedCrossRef Rog DJ, Nurmikko TJ, Young CA (2007) Oromucosal delta9-tetrahydrocannabinol/cannabidiol for neuropathic pain associated with multiple sclerosis: an uncontrolled, open-label, 2-year extension trial. Clin Ther 29:2068–2079PubMedCrossRef
173.
go back to reference Rog DJ, Nurmikko TJ, Friede T, Young CA (2005) Randomized, controlled trial of cannabis-based medicine in central pain in multiple sclerosis. Neurology 65:812–819PubMedCrossRef Rog DJ, Nurmikko TJ, Friede T, Young CA (2005) Randomized, controlled trial of cannabis-based medicine in central pain in multiple sclerosis. Neurology 65:812–819PubMedCrossRef
174.
go back to reference Blake DR, Robson P, Ho M, Jubb RW, McCabe CS (2006) Preliminary assessment of the efficacy, tolerability and safety of a cannabis-based medicine (Sativex) in the treatment of pain caused by rheumatoid arthritis. Rheumatology (Oxford) 45:50–52CrossRef Blake DR, Robson P, Ho M, Jubb RW, McCabe CS (2006) Preliminary assessment of the efficacy, tolerability and safety of a cannabis-based medicine (Sativex) in the treatment of pain caused by rheumatoid arthritis. Rheumatology (Oxford) 45:50–52CrossRef
175.
go back to reference Serpell MG, Notcutt W, Collin C (2013) Sativex long-term use: an open-label trial in patients with spasticity due to multiple sclerosis. J Neurol 260:285–295PubMedCrossRef Serpell MG, Notcutt W, Collin C (2013) Sativex long-term use: an open-label trial in patients with spasticity due to multiple sclerosis. J Neurol 260:285–295PubMedCrossRef
176.
go back to reference Wade DT, Makela PM, House H, Bateman C, Robson P (2006) Long-term use of a cannabis-based medicine in the treatment of spasticity and other symptoms in multiple sclerosis. Mult Scler 12:639–645PubMedCrossRef Wade DT, Makela PM, House H, Bateman C, Robson P (2006) Long-term use of a cannabis-based medicine in the treatment of spasticity and other symptoms in multiple sclerosis. Mult Scler 12:639–645PubMedCrossRef
177.
go back to reference Portenoy RK, Ganae-Motan ED, Allende S et al (2012) Nabiximols for opioid-treated cancer patients with poorly-controlled chronic pain: a randomized, placebo-controlled, graded-dose trial. J Pain 13:438–449PubMedCrossRef Portenoy RK, Ganae-Motan ED, Allende S et al (2012) Nabiximols for opioid-treated cancer patients with poorly-controlled chronic pain: a randomized, placebo-controlled, graded-dose trial. J Pain 13:438–449PubMedCrossRef
178.
go back to reference Wade DT, Robson P, House H, Makela P, Aram J (2003) A preliminary controlled study to determine whether whole-plant cannabis extracts can improve intractable neurogenic symptoms. Clin Rehabil 17:21–29PubMedCrossRef Wade DT, Robson P, House H, Makela P, Aram J (2003) A preliminary controlled study to determine whether whole-plant cannabis extracts can improve intractable neurogenic symptoms. Clin Rehabil 17:21–29PubMedCrossRef
179.
go back to reference Notcutt W, Price M, Miller R et al (2004) Initial experiences with medicinal extracts of cannabis for chronic pain: results from 34 ′N of 1′ studies. Anaesthesia 59:440–452PubMedCrossRef Notcutt W, Price M, Miller R et al (2004) Initial experiences with medicinal extracts of cannabis for chronic pain: results from 34 ′N of 1′ studies. Anaesthesia 59:440–452PubMedCrossRef
180.
go back to reference Notcutt W, Langford R, Davies P, Ratcliffe S, Potts R (2012) 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 18:219–228PubMedCrossRef Notcutt W, Langford R, Davies P, Ratcliffe S, Potts R (2012) 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 18:219–228PubMedCrossRef
181.
go back to reference Berman JS, Symonds C, Birch R (2004) Efficacy of two cannabis based medicinal extracts for relief of central neuropathic pain from brachial plexus avulsion: results of a randomised controlled trial. Pain 112:299–306PubMedCrossRef Berman JS, Symonds C, Birch R (2004) Efficacy of two cannabis based medicinal extracts for relief of central neuropathic pain from brachial plexus avulsion: results of a randomised controlled trial. Pain 112:299–306PubMedCrossRef
182.
go back to reference Langford RM, Mares J, Novotna A et al (2013) 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 260:984–997PubMedCrossRef Langford RM, Mares J, Novotna A et al (2013) 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 260:984–997PubMedCrossRef
183.
go back to reference Johnson JR, Burnell-Nugent M, Lossignol D, Ganae-Motan ED, Potts R, Fallon MT (2010) 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 39:167–179CrossRef Johnson JR, Burnell-Nugent M, Lossignol D, Ganae-Motan ED, Potts R, Fallon MT (2010) 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 39:167–179CrossRef
184.
go back to reference Johnson JR, Lossignol D, Burnell-Nugent M, Fallon MT (2013) 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 46:207–218CrossRef Johnson JR, Lossignol D, Burnell-Nugent M, Fallon MT (2013) 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 46:207–218CrossRef
185.
go back to reference Zajicek JP, Sanders HP, Wright DE et al (2005) Cannabinoids in multiple sclerosis (CAMS) study: safety and efficacy data for 12 months follow up. J Neurol Neurosurg Psychiatry 76:1664–1669PubMedPubMedCentralCrossRef Zajicek JP, Sanders HP, Wright DE et al (2005) Cannabinoids in multiple sclerosis (CAMS) study: safety and efficacy data for 12 months follow up. J Neurol Neurosurg Psychiatry 76:1664–1669PubMedPubMedCentralCrossRef
186.
187.
go back to reference Zajicek JP, Hobart JC, Slade A, Barnes D, Mattison PG, MUSEC Research Group (2012) Multiple sclerosis and extract of cannabis: results of the MUSEC trial. J Neurol Neurosurg Psychiatry 83:1125–1132PubMedCrossRef Zajicek JP, Hobart JC, Slade A, Barnes D, Mattison PG, MUSEC Research Group (2012) Multiple sclerosis and extract of cannabis: results of the MUSEC trial. J Neurol Neurosurg Psychiatry 83:1125–1132PubMedCrossRef
188.
go back to reference Zajicek J, Fox P, Sanders H et al (2003) Cannabinoids for treatment of spasticity and other symptoms related to multiple sclerosis (CAMS study): multicentre randomised placebo-controlled trial. Lancet 362:1517–1526PubMedCrossRef Zajicek J, Fox P, Sanders H et al (2003) Cannabinoids for treatment of spasticity and other symptoms related to multiple sclerosis (CAMS study): multicentre randomised placebo-controlled trial. Lancet 362:1517–1526PubMedCrossRef
189.
go back to reference Vaney C, Heinzel-Gutenbrunner M, Jobin P et al (2004) 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 10:417–424PubMedCrossRef Vaney C, Heinzel-Gutenbrunner M, Jobin P et al (2004) 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 10:417–424PubMedCrossRef
190.
go back to reference Collin C, Davies P, Mutiboko IK, Ratcliffe S, Sativex Spasticity in MS Study Group (2007) Randomized controlled trial of cannabis-based medicine in spasticity caused by multiple sclerosis. Eur J Neurol 14:290–296PubMedCrossRef Collin C, Davies P, Mutiboko IK, Ratcliffe S, Sativex Spasticity in MS Study Group (2007) Randomized controlled trial of cannabis-based medicine in spasticity caused by multiple sclerosis. Eur J Neurol 14:290–296PubMedCrossRef
191.
go back to reference Collin C, Ehler E, Waberzinek G et al (2010) A double-blind, randomized, placebo-controlled, parallel-group study of Sativex, in subjects with symptoms of spasticity due to multiple sclerosis. Neurol Res 32:451–459PubMedCrossRef Collin C, Ehler E, Waberzinek G et al (2010) A double-blind, randomized, placebo-controlled, parallel-group study of Sativex, in subjects with symptoms of spasticity due to multiple sclerosis. Neurol Res 32:451–459PubMedCrossRef
192.
go back to reference Wade DT, Makela P, Robson P, House H, Bateman C (2004) 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 10:434–441PubMedCrossRef Wade DT, Makela P, Robson P, House H, Bateman C (2004) 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 10:434–441PubMedCrossRef
193.
go back to reference Serpell M, Ratcliffe S, Hovorka J et al (2014) A double-blind, randomized, placebo-controlled, parallel group study of THC/CBD spray in peripheral neuropathic pain treatment. Eur J Pain 18:999–1012PubMedCrossRef Serpell M, Ratcliffe S, Hovorka J et al (2014) A double-blind, randomized, placebo-controlled, parallel group study of THC/CBD spray in peripheral neuropathic pain treatment. Eur J Pain 18:999–1012PubMedCrossRef
194.
go back to reference Syed YY, McKeage K, Scott LJ (2014) Delta-9-tetrahydrocannabinol/cannabidiol (Sativex(R)): a review of its use in patients with moderate to severe spasticity due to multiple sclerosis. Drugs 74:563–578PubMedCrossRef Syed YY, McKeage K, Scott LJ (2014) Delta-9-tetrahydrocannabinol/cannabidiol (Sativex(R)): a review of its use in patients with moderate to severe spasticity due to multiple sclerosis. Drugs 74:563–578PubMedCrossRef
195.
196.
go back to reference Barnes MP (2006) Sativex: clinical efficacy and tolerability in the treatment of symptoms of multiple sclerosis and neuropathic pain. Expert Opin Pharmacother 7:607–615PubMedCrossRef Barnes MP (2006) Sativex: clinical efficacy and tolerability in the treatment of symptoms of multiple sclerosis and neuropathic pain. Expert Opin Pharmacother 7:607–615PubMedCrossRef
197.
go back to reference GW Pharmaceuticals. Sativex product monograph., 2010 GW Pharmaceuticals. Sativex product monograph., 2010
198.
go back to reference Holdcroft A, Maze M, Dore C, Tebbs S, Thompson S (2006) A multicenter dose-escalation study of the analgesic and adverse effects of an oral cannabis extract (Cannador) for postoperative pain management. Anesthesiology 104:1040–1046PubMedCrossRef Holdcroft A, Maze M, Dore C, Tebbs S, Thompson S (2006) A multicenter dose-escalation study of the analgesic and adverse effects of an oral cannabis extract (Cannador) for postoperative pain management. Anesthesiology 104:1040–1046PubMedCrossRef
199.
go back to reference Sallan SE, Zinberg NE, Frei E 3rd (1975) Antiemetic effect of delta-9-tetrahydrocannabinol in patients receiving cancer chemotherapy. N Engl J Med 293:795–797PubMedCrossRef Sallan SE, Zinberg NE, Frei E 3rd (1975) Antiemetic effect of delta-9-tetrahydrocannabinol in patients receiving cancer chemotherapy. N Engl J Med 293:795–797PubMedCrossRef
200.
go back to reference Sallan SE, Cronin C, Zelen M, Zinberg NE (1980) Antiemetics in patients receiving chemotherapy for cancer: a randomized comparison of delta-9-tetrahydrocannabinol and prochlorperazine. N Engl J Med 302:135–138PubMedCrossRef Sallan SE, Cronin C, Zelen M, Zinberg NE (1980) Antiemetics in patients receiving chemotherapy for cancer: a randomized comparison of delta-9-tetrahydrocannabinol and prochlorperazine. N Engl J Med 302:135–138PubMedCrossRef
201.
go back to reference Vinciguerra V, Moore T, Brennan E (1988) Inhalation marijuana as an antiemetic for cancer chemotherapy. N Y State J Med 88:525–527PubMed Vinciguerra V, Moore T, Brennan E (1988) Inhalation marijuana as an antiemetic for cancer chemotherapy. N Y State J Med 88:525–527PubMed
202.
go back to reference Carey MP, Burish TG, Brenner DE (1983) Delta-9-tetrahydrocannabinol in cancer chemotherapy: research problems and issues. Ann Intern Med 99:106–114PubMedCrossRef Carey MP, Burish TG, Brenner DE (1983) Delta-9-tetrahydrocannabinol in cancer chemotherapy: research problems and issues. Ann Intern Med 99:106–114PubMedCrossRef
203.
go back to reference Lucas VS Jr, Laszlo J (1980) delta 9-tetrahydrocannabinol for refractory vomiting induced by cancer chemotherapy. JAMA 243:1241–1243PubMedCrossRef Lucas VS Jr, Laszlo J (1980) delta 9-tetrahydrocannabinol for refractory vomiting induced by cancer chemotherapy. JAMA 243:1241–1243PubMedCrossRef
204.
go back to reference Frytak S, Moertel CG, O'Fallon JR et al (1979) Delta-9-tetrahydrocannabinol as an antiemetic for patients receiving cancer chemotherapy. A comparison with prochlorperazine and a placebo. Ann Intern Med 91:825–830PubMedCrossRef Frytak S, Moertel CG, O'Fallon JR et al (1979) Delta-9-tetrahydrocannabinol as an antiemetic for patients receiving cancer chemotherapy. A comparison with prochlorperazine and a placebo. Ann Intern Med 91:825–830PubMedCrossRef
205.
go back to reference Ungerleider JT, Andrysiak T, Fairbanks L, Goodnight J, Sarna G, Jamison K (1982) Cannabis and cancer chemotherapy: a comparison of oral delta-9-THC and prochlorperazine. Cancer 50:636–645PubMedCrossRef Ungerleider JT, Andrysiak T, Fairbanks L, Goodnight J, Sarna G, Jamison K (1982) Cannabis and cancer chemotherapy: a comparison of oral delta-9-THC and prochlorperazine. Cancer 50:636–645PubMedCrossRef
206.
go back to reference Orr LE, McKernan JF, Bloome B (1980) Antiemetic effect of tetrahydrocannabinol. Compared with placebo and prochlorperazine in chemotherapy-associated nausea and emesis. Arch Intern Med 140:1431–1433PubMedCrossRef Orr LE, McKernan JF, Bloome B (1980) Antiemetic effect of tetrahydrocannabinol. Compared with placebo and prochlorperazine in chemotherapy-associated nausea and emesis. Arch Intern Med 140:1431–1433PubMedCrossRef
207.
go back to reference Orr LE, McKernan JF (1981) Antiemetic effect of delta 9-tetrahydrocannabinol in chemotherapy-associated nausea and emesis as compared to placebo and compazine. J Clin Pharmacol 21:76S–80SPubMedCrossRef Orr LE, McKernan JF (1981) Antiemetic effect of delta 9-tetrahydrocannabinol in chemotherapy-associated nausea and emesis as compared to placebo and compazine. J Clin Pharmacol 21:76S–80SPubMedCrossRef
208.
go back to reference Levitt M, Wilson A, Bowman D et al (1981) Physiologic observations in a controlled clinical trial of the antiemetic effectiveness of 5, 10, and 15 mg of delta 9-tetrahydrocannabinol in cancer chemotherapy. Ophthalmologic implications. J Clin Pharmacol 21:103S–109SPubMedCrossRef Levitt M, Wilson A, Bowman D et al (1981) Physiologic observations in a controlled clinical trial of the antiemetic effectiveness of 5, 10, and 15 mg of delta 9-tetrahydrocannabinol in cancer chemotherapy. Ophthalmologic implications. J Clin Pharmacol 21:103S–109SPubMedCrossRef
209.
go back to reference McCabe M, Smith FP, Macdonald JS, Woolley PV, Goldberg D, Schein PS (1988) Efficacy of tetrahydrocannabinol in patients refractory to standard antiemetic therapy. Investig New Drugs 6:243–246CrossRef McCabe M, Smith FP, Macdonald JS, Woolley PV, Goldberg D, Schein PS (1988) Efficacy of tetrahydrocannabinol in patients refractory to standard antiemetic therapy. Investig New Drugs 6:243–246CrossRef
210.
go back to reference Niiranen A, Mattson K (1985) A cross-over comparison of nabilone and prochlorperazine for emesis induced by cancer chemotherapy. Am J Clin Oncol 8:336–340PubMedCrossRef Niiranen A, Mattson K (1985) A cross-over comparison of nabilone and prochlorperazine for emesis induced by cancer chemotherapy. Am J Clin Oncol 8:336–340PubMedCrossRef
211.
go back to reference Herman TS, Einhorn LH, Jones SE et al (1979) Superiority of nabilone over prochlorperazine as an antiemetic in patients receiving cancer chemotherapy. N Engl J Med 300:1295–1297PubMedCrossRef Herman TS, Einhorn LH, Jones SE et al (1979) Superiority of nabilone over prochlorperazine as an antiemetic in patients receiving cancer chemotherapy. N Engl J Med 300:1295–1297PubMedCrossRef
212.
go back to reference Vincent BJ, McQuiston DJ, Einhorn LH, Nagy CM, Brames MJ (1983) Review of cannabinoids and their antiemetic effectiveness. Drugs 25(Suppl 1):52–62PubMedCrossRef Vincent BJ, McQuiston DJ, Einhorn LH, Nagy CM, Brames MJ (1983) Review of cannabinoids and their antiemetic effectiveness. Drugs 25(Suppl 1):52–62PubMedCrossRef
213.
go back to reference Einhorn L (1982) Nabilone: an effective antiemetic agent in patients receiving cancer chemotherapy. Cancer Treat Rev 9(Suppl B):55–61PubMedCrossRef Einhorn L (1982) Nabilone: an effective antiemetic agent in patients receiving cancer chemotherapy. Cancer Treat Rev 9(Suppl B):55–61PubMedCrossRef
214.
go back to reference Einhorn LH, Nagy C, Furnas B, Williams SD (1981) Nabilone: an effective antiemetic in patients receiving cancer chemotherapy. J Clin Pharmacol 21:64S–69SPubMedCrossRef Einhorn LH, Nagy C, Furnas B, Williams SD (1981) Nabilone: an effective antiemetic in patients receiving cancer chemotherapy. J Clin Pharmacol 21:64S–69SPubMedCrossRef
215.
go back to reference Ahmedzai S, Carlyle DL, Calder IT, Moran F (1983) Anti-emetic efficacy and toxicity of nabilone, a synthetic cannabinoid, in lung cancer chemotherapy. Br J Cancer 48:657–663PubMedPubMedCentralCrossRef Ahmedzai S, Carlyle DL, Calder IT, Moran F (1983) Anti-emetic efficacy and toxicity of nabilone, a synthetic cannabinoid, in lung cancer chemotherapy. Br J Cancer 48:657–663PubMedPubMedCentralCrossRef
216.
go back to reference Garb S, Beers AL, Bograd M et al (1980) Two-pronged study of tetrahydrocannabinol (THC) prevention of vomiting for cancer chemotherapy. IRCS Med Sci 8:203–204 Garb S, Beers AL, Bograd M et al (1980) Two-pronged study of tetrahydrocannabinol (THC) prevention of vomiting for cancer chemotherapy. IRCS Med Sci 8:203–204
217.
go back to reference Lane M, Smith FE, Sullivan RA, Plasse TF (1990) Dronabinol and prochlorperazine alone and in combination as antiemetic agents for cancer chemotherapy. Am J Clin Oncol 13:480–484PubMedCrossRef Lane M, Smith FE, Sullivan RA, Plasse TF (1990) Dronabinol and prochlorperazine alone and in combination as antiemetic agents for cancer chemotherapy. Am J Clin Oncol 13:480–484PubMedCrossRef
218.
go back to reference Lane M, Vogel CL, Ferguson J et al (1991) Dronabinol and prochlorperazine in combination for treatment of cancer chemotherapy-induced nausea and vomiting. J Pain Symptom Manag 6:352–359CrossRef Lane M, Vogel CL, Ferguson J et al (1991) Dronabinol and prochlorperazine in combination for treatment of cancer chemotherapy-induced nausea and vomiting. J Pain Symptom Manag 6:352–359CrossRef
219.
go back to reference Plasse TF, Gorter RW, Krasnow SH, Lane M, Shepard KV, Wadleigh RG (1991) Recent clinical experience with dronabinol. Pharmacol Biochem Behav 40:695–700PubMedCrossRef Plasse TF, Gorter RW, Krasnow SH, Lane M, Shepard KV, Wadleigh RG (1991) Recent clinical experience with dronabinol. Pharmacol Biochem Behav 40:695–700PubMedCrossRef
220.
go back to reference Cunningham D, Forrest GJ, Soukop M, Gilchrist NL, Calder IT, McArdle CS (1985) Nabilone and prochlorperazine: a useful combination for emesis induced by cytotoxic drugs. Br Med J (Clin Res Ed) 291:864–865CrossRef Cunningham D, Forrest GJ, Soukop M, Gilchrist NL, Calder IT, McArdle CS (1985) Nabilone and prochlorperazine: a useful combination for emesis induced by cytotoxic drugs. Br Med J (Clin Res Ed) 291:864–865CrossRef
221.
go back to reference Gonzalez-Rosales F, Walsh D (1997) Intractable nausea and vomiting due to gastrointestinal mucosal metastases relieved by tetrahydrocannabinol (dronabinol). J Pain Symptom Manag 14:311–314CrossRef Gonzalez-Rosales F, Walsh D (1997) Intractable nausea and vomiting due to gastrointestinal mucosal metastases relieved by tetrahydrocannabinol (dronabinol). J Pain Symptom Manag 14:311–314CrossRef
222.
go back to reference Chang AE, Shiling DJ, Stillman RC et al (1979) Delata-9-tetrahydrocannabinol as an antiemetic in cancer patients receiving high-dose methotrexate. A prospective, randomized evaluation. Ann Intern Med 91:819–824PubMedCrossRef Chang AE, Shiling DJ, Stillman RC et al (1979) Delata-9-tetrahydrocannabinol as an antiemetic in cancer patients receiving high-dose methotrexate. A prospective, randomized evaluation. Ann Intern Med 91:819–824PubMedCrossRef
223.
go back to reference Staquet M, Bron D, Rozencweig M, Kenis Y (1981) Clinical studies with a THC analog (BRL-4664) in the prevention of cisplatin-induced vomiting. J Clin Pharmacol 21:60S–63SPubMedCrossRef Staquet M, Bron D, Rozencweig M, Kenis Y (1981) Clinical studies with a THC analog (BRL-4664) in the prevention of cisplatin-induced vomiting. J Clin Pharmacol 21:60S–63SPubMedCrossRef
224.
go back to reference Sharkey KA, Darmani NA, Parker LA (2014) Regulation of nausea and vomiting by cannabinoids and the endocannabinoid system. Eur J Pharmacol 722:134–146PubMedCrossRef Sharkey KA, Darmani NA, Parker LA (2014) Regulation of nausea and vomiting by cannabinoids and the endocannabinoid system. Eur J Pharmacol 722:134–146PubMedCrossRef
226.
go back to reference Tramer MR, Carroll D, Campbell FA, Reynolds DJ, Moore RA, McQuay HJ (2001) Cannabinoids for control of chemotherapy induced nausea and vomiting: quantitative systematic review. BMJ 323:16–21PubMedPubMedCentralCrossRef Tramer MR, Carroll D, Campbell FA, Reynolds DJ, Moore RA, McQuay HJ (2001) Cannabinoids for control of chemotherapy induced nausea and vomiting: quantitative systematic review. BMJ 323:16–21PubMedPubMedCentralCrossRef
227.
go back to reference Sutton IR, Daeninck P (2006) Cannabinoids in the management of intractable chemotherapy-induced nausea and vomiting and cancer-related pain. J Support Oncol 4:531–535PubMed Sutton IR, Daeninck P (2006) Cannabinoids in the management of intractable chemotherapy-induced nausea and vomiting and cancer-related pain. J Support Oncol 4:531–535PubMed
228.
go back to reference Pisanti S, Malfitano AM, Grimaldi C et al (2009) Use of cannabinoid receptor agonists in cancer therapy as palliative and curative agents. Best Pract Res Clin Endocrinol Metab 23:117–131PubMedCrossRef Pisanti S, Malfitano AM, Grimaldi C et al (2009) Use of cannabinoid receptor agonists in cancer therapy as palliative and curative agents. Best Pract Res Clin Endocrinol Metab 23:117–131PubMedCrossRef
229.
go back to reference Tortorice PV, O'Connell MB (1990) Management of chemotherapy-induced nausea and vomiting. Pharmacotherapy 10:129–145PubMed Tortorice PV, O'Connell MB (1990) Management of chemotherapy-induced nausea and vomiting. Pharmacotherapy 10:129–145PubMed
230.
go back to reference Cunningham D, Bradley CJ, Forrest GJ et al (1988) A randomized trial of oral nabilone and prochlorperazine compared to intravenous metoclopramide and dexamethasone in the treatment of nausea and vomiting induced by chemotherapy regimens containing cisplatin or cisplatin analogues. Eur J Cancer Clin Oncol 24:685–689PubMedCrossRef Cunningham D, Bradley CJ, Forrest GJ et al (1988) A randomized trial of oral nabilone and prochlorperazine compared to intravenous metoclopramide and dexamethasone in the treatment of nausea and vomiting induced by chemotherapy regimens containing cisplatin or cisplatin analogues. Eur J Cancer Clin Oncol 24:685–689PubMedCrossRef
231.
go back to reference Soderpalm AH, Schuster A, de Wit H (2001) Antiemetic efficacy of smoked marijuana: subjective and behavioral effects on nausea induced by syrup of ipecac. Pharmacol Biochem Behav 69:343–350PubMedCrossRef Soderpalm AH, Schuster A, de Wit H (2001) Antiemetic efficacy of smoked marijuana: subjective and behavioral effects on nausea induced by syrup of ipecac. Pharmacol Biochem Behav 69:343–350PubMedCrossRef
232.
go back to reference Machado Rocha FC, Stefano SC, De Cassia Haiek R, Rosa Oliveira LM, Da Silveira DX (2008) Therapeutic use of cannabis sativa on chemotherapy-induced nausea and vomiting among cancer patients: systematic review and meta-analysis. Eur J Cancer Care (Engl) 17:431–443CrossRef Machado Rocha FC, Stefano SC, De Cassia Haiek R, Rosa Oliveira LM, Da Silveira DX (2008) Therapeutic use of cannabis sativa on chemotherapy-induced nausea and vomiting among cancer patients: systematic review and meta-analysis. Eur J Cancer Care (Engl) 17:431–443CrossRef
233.
go back to reference Musty R, Rossi R (2001) Effects of smoked cannabis and oral delta-9-tetrahydrocannabinol on nausea and emesis after cancer chemotherapy: a review of state clinical trials. J Cannabis Ther 1:29–42CrossRef Musty R, Rossi R (2001) Effects of smoked cannabis and oral delta-9-tetrahydrocannabinol on nausea and emesis after cancer chemotherapy: a review of state clinical trials. J Cannabis Ther 1:29–42CrossRef
234.
go back to reference Meiri E, Jhangiani H, Vredenburgh JJ et al (2007) Efficacy of dronabinol alone and in combination with ondansetron versus ondansetron alone for delayed chemotherapy-induced nausea and vomiting. Curr Med Res Opin 23:533–543PubMedCrossRef Meiri E, Jhangiani H, Vredenburgh JJ et al (2007) Efficacy of dronabinol alone and in combination with ondansetron versus ondansetron alone for delayed chemotherapy-induced nausea and vomiting. Curr Med Res Opin 23:533–543PubMedCrossRef
235.
go back to reference Ekert H, Waters KD, Jurk IH, Mobilia J, Loughnan P (1979) Amelioration of cancer chemotherapy-induced nausea and vomiting by delta-9-tetrahydrocannabinol. Med J Aust 2:657–659PubMed Ekert H, Waters KD, Jurk IH, Mobilia J, Loughnan P (1979) Amelioration of cancer chemotherapy-induced nausea and vomiting by delta-9-tetrahydrocannabinol. Med J Aust 2:657–659PubMed
236.
go back to reference Pertwee RG (2012) Targeting the endocannabinoid system with cannabinoid receptor agonists: pharmacological strategies and therapeutic possibilities. Philos Trans R Soc Lond Ser B Biol Sci 367:3353–3363CrossRef Pertwee RG (2012) Targeting the endocannabinoid system with cannabinoid receptor agonists: pharmacological strategies and therapeutic possibilities. Philos Trans R Soc Lond Ser B Biol Sci 367:3353–3363CrossRef
237.
go back to reference Rock EM, Connolly C, Limebeer CL, Parker LA (2016) Effect of combined oral doses of Delta(9)-tetrahydrocannabinol (THC) and cannabidiolic acid (CBDA) on acute and anticipatory nausea in rat models. Psychopharmacology 233:3353–3360PubMedCrossRef Rock EM, Connolly C, Limebeer CL, Parker LA (2016) Effect of combined oral doses of Delta(9)-tetrahydrocannabinol (THC) and cannabidiolic acid (CBDA) on acute and anticipatory nausea in rat models. Psychopharmacology 233:3353–3360PubMedCrossRef
238.
go back to reference Green ST, Nathwani D, Goldberg DJ, Kennedy DH (1989) Nabilone as effective therapy for intractable nausea and vomiting in AIDS. Br J Clin Pharmacol 28:494–495PubMedPubMedCentralCrossRef Green ST, Nathwani D, Goldberg DJ, Kennedy DH (1989) Nabilone as effective therapy for intractable nausea and vomiting in AIDS. Br J Clin Pharmacol 28:494–495PubMedPubMedCentralCrossRef
239.
go back to reference Chan HS, Correia JA, MacLeod SM (1987) Nabilone versus prochlorperazine for control of cancer chemotherapy-induced emesis in children: a double-blind, crossover trial. Pediatrics 79:946–952PubMed Chan HS, Correia JA, MacLeod SM (1987) Nabilone versus prochlorperazine for control of cancer chemotherapy-induced emesis in children: a double-blind, crossover trial. Pediatrics 79:946–952PubMed
241.
go back to reference Abrahamov A, Abrahamov A, Mechoulam R (1995) An efficient new cannabinoid antiemetic in pediatric oncology. Life Sci 56:2097–2102PubMedCrossRef Abrahamov A, Abrahamov A, Mechoulam R (1995) An efficient new cannabinoid antiemetic in pediatric oncology. Life Sci 56:2097–2102PubMedCrossRef
242.
go back to reference Abbott Products Inc. Marinol product monograph., 2010 Abbott Products Inc. Marinol product monograph., 2010
243.
go back to reference Valeant Canada. Cesamet product monograph., 2009 Valeant Canada. Cesamet product monograph., 2009
244.
go back to reference Narang S, Gibson D, Wasan AD et al (2008) Efficacy of dronabinol as an adjuvant treatment for chronic pain patients on opioid therapy. J Pain 9:254–264PubMedCrossRef Narang S, Gibson D, Wasan AD et al (2008) Efficacy of dronabinol as an adjuvant treatment for chronic pain patients on opioid therapy. J Pain 9:254–264PubMedCrossRef
245.
go back to reference Svendsen KB, Jensen TS, Bach FW (2005) Effect of the synthetic cannabinoid dronabinol on central pain in patients with multiple sclerosis--secondary publication. Ugeskr Laeger 167:2772–2774PubMed Svendsen KB, Jensen TS, Bach FW (2005) Effect of the synthetic cannabinoid dronabinol on central pain in patients with multiple sclerosis--secondary publication. Ugeskr Laeger 167:2772–2774PubMed
246.
go back to reference Svendsen KB, Jensen TS, Bach FW (2004) Does the cannabinoid dronabinol reduce central pain in multiple sclerosis? Randomised double blind placebo controlled crossover trial. BMJ 329:253PubMedPubMedCentralCrossRef Svendsen KB, Jensen TS, Bach FW (2004) Does the cannabinoid dronabinol reduce central pain in multiple sclerosis? Randomised double blind placebo controlled crossover trial. BMJ 329:253PubMedPubMedCentralCrossRef
247.
go back to reference Skrabek RQ, Galimova L, Ethans K, Perry D (2008) Nabilone for the treatment of pain in fibromyalgia. J Pain 9:164–173PubMedCrossRef Skrabek RQ, Galimova L, Ethans K, Perry D (2008) Nabilone for the treatment of pain in fibromyalgia. J Pain 9:164–173PubMedCrossRef
248.
go back to reference Naef M, Curatolo M, Petersen-Felix S, Arendt-Nielsen L, Zbinden A, Brenneisen R (2003) The analgesic effect of oral delta-9-tetrahydrocannabinol (THC), morphine, and a THC-morphine combination in healthy subjects under experimental pain conditions. Pain 105:79–88PubMedCrossRef Naef M, Curatolo M, Petersen-Felix S, Arendt-Nielsen L, Zbinden A, Brenneisen R (2003) The analgesic effect of oral delta-9-tetrahydrocannabinol (THC), morphine, and a THC-morphine combination in healthy subjects under experimental pain conditions. Pain 105:79–88PubMedCrossRef
249.
go back to reference Pinsger M, Schimetta W, Volc D, Hiermann E, Riederer F, Polz W (2006) Benefits of an add-on treatment with the synthetic cannabinomimetic nabilone on patients with chronic pain--a randomized controlled trial. Wien Klin Wochenschr 118:327–335PubMedCrossRef Pinsger M, Schimetta W, Volc D, Hiermann E, Riederer F, Polz W (2006) Benefits of an add-on treatment with the synthetic cannabinomimetic nabilone on patients with chronic pain--a randomized controlled trial. Wien Klin Wochenschr 118:327–335PubMedCrossRef
250.
go back to reference Wissel J, Haydn T, Muller J et al (2006) Low dose treatment with the synthetic cannabinoid Nabilone significantly reduces spasticity-related pain : a double-blind placebo-controlled cross-over trial. J Neurol 253:1337–1341PubMedCrossRef Wissel J, Haydn T, Muller J et al (2006) Low dose treatment with the synthetic cannabinoid Nabilone significantly reduces spasticity-related pain : a double-blind placebo-controlled cross-over trial. J Neurol 253:1337–1341PubMedCrossRef
251.
go back to reference Toth C, Mawani S, Brady S et al (2012) An enriched-enrolment, randomized withdrawal, flexible-dose, double-blind, placebo-controlled, parallel assignment efficacy study of nabilone as adjuvant in the treatment of diabetic peripheral neuropathic pain. Pain 153:2073–2082PubMedCrossRef Toth C, Mawani S, Brady S et al (2012) An enriched-enrolment, randomized withdrawal, flexible-dose, double-blind, placebo-controlled, parallel assignment efficacy study of nabilone as adjuvant in the treatment of diabetic peripheral neuropathic pain. Pain 153:2073–2082PubMedCrossRef
252.
go back to reference Turcotte D, Doupe M, Torabi M et al (2015) Nabilone as an adjunctive to gabapentin for multiple sclerosis-induced neuropathic pain: a randomized controlled trial. Pain Med 16:149–159PubMedCrossRef Turcotte D, Doupe M, Torabi M et al (2015) Nabilone as an adjunctive to gabapentin for multiple sclerosis-induced neuropathic pain: a randomized controlled trial. Pain Med 16:149–159PubMedCrossRef
253.
go back to reference Notcutt WG, Price M, Chapman G (1997) Clinical experience with nabilone for chronic pain. Pharm Sci 3:551–555 Notcutt WG, Price M, Chapman G (1997) Clinical experience with nabilone for chronic pain. Pharm Sci 3:551–555
254.
go back to reference Hamann W, di Vadi PP (1999) Analgesic effect of the cannabinoid analogue nabilone is not mediated by opioid receptors. Lancet 353:560PubMedCrossRef Hamann W, di Vadi PP (1999) Analgesic effect of the cannabinoid analogue nabilone is not mediated by opioid receptors. Lancet 353:560PubMedCrossRef
255.
go back to reference Berlach DM, Shir Y, Ware MA (2006) Experience with the synthetic cannabinoid nabilone in chronic noncancer pain. Pain Med 7:25–29PubMedCrossRef Berlach DM, Shir Y, Ware MA (2006) Experience with the synthetic cannabinoid nabilone in chronic noncancer pain. Pain Med 7:25–29PubMedCrossRef
256.
259.
go back to reference Zuardi AW (2008) Cannabidiol: from an inactive cannabinoid to a drug with wide spectrum of action. Rev Bras Psiquiatr 30:271–280PubMedCrossRef Zuardi AW (2008) Cannabidiol: from an inactive cannabinoid to a drug with wide spectrum of action. Rev Bras Psiquiatr 30:271–280PubMedCrossRef
260.
go back to reference Mechoulam R, Parker LA, Gallily R (2002) Cannabidiol: an overview of some pharmacological aspects. J Clin Pharmacol 42:11S–19SPubMedCrossRef Mechoulam R, Parker LA, Gallily R (2002) Cannabidiol: an overview of some pharmacological aspects. J Clin Pharmacol 42:11S–19SPubMedCrossRef
261.
go back to reference Pagano E, Capasso R, Piscitelli F et al (2016) An orally active Cannabis extract with high content in cannabidiol attenuates chemically-induced intestinal inflammation and hypermotility in the mouse. Front Pharmacol 7:341PubMedPubMedCentralCrossRef Pagano E, Capasso R, Piscitelli F et al (2016) An orally active Cannabis extract with high content in cannabidiol attenuates chemically-induced intestinal inflammation and hypermotility in the mouse. Front Pharmacol 7:341PubMedPubMedCentralCrossRef
262.
go back to reference Mechoulam R, Peters M, Murillo-Rodriguez E, Hanus LO (2007) Cannabidiol--recent advances. Chem Biodivers 4:1678–1692PubMedCrossRef Mechoulam R, Peters M, Murillo-Rodriguez E, Hanus LO (2007) Cannabidiol--recent advances. Chem Biodivers 4:1678–1692PubMedCrossRef
263.
go back to reference Malfait AM, Gallily R, Sumariwalla PF et al (2000) The nonpsychoactive cannabis constituent cannabidiol is an oral anti-arthritic therapeutic in murine collagen-induced arthritis. Proc Natl Acad Sci U S A 97:9561–9566PubMedPubMedCentralCrossRef Malfait AM, Gallily R, Sumariwalla PF et al (2000) The nonpsychoactive cannabis constituent cannabidiol is an oral anti-arthritic therapeutic in murine collagen-induced arthritis. Proc Natl Acad Sci U S A 97:9561–9566PubMedPubMedCentralCrossRef
264.
go back to reference Formukong EA, Evans AT, Evans FJ (1988) Analgesic and antiinflammatory activity of constituents of cannabis sativa L. Inflammation 12:361–371PubMedCrossRef Formukong EA, Evans AT, Evans FJ (1988) Analgesic and antiinflammatory activity of constituents of cannabis sativa L. Inflammation 12:361–371PubMedCrossRef
265.
go back to reference Formukong EA, Evans AT, Evans FJ (1989) The medicinal uses of cannabis and its constituents. Phytother Res 3:219–231CrossRef Formukong EA, Evans AT, Evans FJ (1989) The medicinal uses of cannabis and its constituents. Phytother Res 3:219–231CrossRef
266.
go back to reference Costa B, Trovato AE, Comelli F, Giagnoni G, Colleoni M (2007) The non-psychoactive cannabis constituent cannabidiol is an orally effective therapeutic agent in rat chronic inflammatory and neuropathic pain. Eur J Pharmacol 556:75–83PubMedCrossRef Costa B, Trovato AE, Comelli F, Giagnoni G, Colleoni M (2007) The non-psychoactive cannabis constituent cannabidiol is an orally effective therapeutic agent in rat chronic inflammatory and neuropathic pain. Eur J Pharmacol 556:75–83PubMedCrossRef
267.
go back to reference Costa B, Colleoni M, Conti S et al (2004) Oral anti-inflammatory activity of cannabidiol, a non-psychoactive constituent of cannabis, in acute carrageenan-induced inflammation in the rat paw. Naunyn Schmiedeberg's Arch Pharmacol 369:294–299CrossRef Costa B, Colleoni M, Conti S et al (2004) Oral anti-inflammatory activity of cannabidiol, a non-psychoactive constituent of cannabis, in acute carrageenan-induced inflammation in the rat paw. Naunyn Schmiedeberg's Arch Pharmacol 369:294–299CrossRef
268.
go back to reference Booz GW (2011) Cannabidiol as an emergent therapeutic strategy for lessening the impact of inflammation on oxidative stress. Free Radic Biol Med 51:1054–1061PubMedPubMedCentralCrossRef Booz GW (2011) Cannabidiol as an emergent therapeutic strategy for lessening the impact of inflammation on oxidative stress. Free Radic Biol Med 51:1054–1061PubMedPubMedCentralCrossRef
269.
go back to reference McHugh D, Tanner C, Mechoulam R, Pertwee RG, Ross RA (2008) Inhibition of human neutrophil chemotaxis by endogenous cannabinoids and phytocannabinoids: evidence for a site distinct from CB1 and CB2. Mol Pharmacol 73:441–450PubMedCrossRef McHugh D, Tanner C, Mechoulam R, Pertwee RG, Ross RA (2008) Inhibition of human neutrophil chemotaxis by endogenous cannabinoids and phytocannabinoids: evidence for a site distinct from CB1 and CB2. Mol Pharmacol 73:441–450PubMedCrossRef
270.
go back to reference Zhornitsky S, Potvin S (2012) Cannabidiol in humans-the quest for therapeutic targets. Pharmaceuticals (Basel) 5:529–552CrossRef Zhornitsky S, Potvin S (2012) Cannabidiol in humans-the quest for therapeutic targets. Pharmaceuticals (Basel) 5:529–552CrossRef
271.
go back to reference Ribeiro A, Almeida VI, Costola-de-Souza C et al (2015) Cannabidiol improves lung function and inflammation in mice submitted to LPS-induced acute lung injury. Immunopharmacol Immunotoxicol 37:35–41PubMedCrossRef Ribeiro A, Almeida VI, Costola-de-Souza C et al (2015) Cannabidiol improves lung function and inflammation in mice submitted to LPS-induced acute lung injury. Immunopharmacol Immunotoxicol 37:35–41PubMedCrossRef
272.
go back to reference Ribeiro A, Ferraz-de-Paula V, Pinheiro ML et al (2012) Cannabidiol, a non-psychotropic plant-derived cannabinoid, decreases inflammation in a murine model of acute lung injury: role for the adenosine A(2A) receptor. Eur J Pharmacol 678:78–85PubMedCrossRef Ribeiro A, Ferraz-de-Paula V, Pinheiro ML et al (2012) Cannabidiol, a non-psychotropic plant-derived cannabinoid, decreases inflammation in a murine model of acute lung injury: role for the adenosine A(2A) receptor. Eur J Pharmacol 678:78–85PubMedCrossRef
273.
go back to reference Kozela E, Lev N, Kaushansky N et al (2011) Cannabidiol inhibits pathogenic T cells, decreases spinal microglial activation and ameliorates multiple sclerosis-like disease in C57BL/6 mice. Br J Pharmacol 163:1507–1519PubMedPubMedCentralCrossRef Kozela E, Lev N, Kaushansky N et al (2011) Cannabidiol inhibits pathogenic T cells, decreases spinal microglial activation and ameliorates multiple sclerosis-like disease in C57BL/6 mice. Br J Pharmacol 163:1507–1519PubMedPubMedCentralCrossRef
274.
go back to reference Mecha M, Feliu A, Inigo PM, Mestre L, Carrillo-Salinas FJ, Guaza C (2013) Cannabidiol provides long-lasting protection against the deleterious effects of inflammation in a viral model of multiple sclerosis: a role for A2A receptors. Neurobiol Dis 59:141–150PubMedCrossRef Mecha M, Feliu A, Inigo PM, Mestre L, Carrillo-Salinas FJ, Guaza C (2013) Cannabidiol provides long-lasting protection against the deleterious effects of inflammation in a viral model of multiple sclerosis: a role for A2A receptors. Neurobiol Dis 59:141–150PubMedCrossRef
275.
go back to reference Mecha M, Torrao AS, Mestre L, Carrillo-Salinas FJ, Mechoulam R, Guaza C (2012) Cannabidiol protects oligodendrocyte progenitor cells from inflammation-induced apoptosis by attenuating endoplasmic reticulum stress. Cell Death Dis 3:e331PubMedPubMedCentralCrossRef Mecha M, Torrao AS, Mestre L, Carrillo-Salinas FJ, Mechoulam R, Guaza C (2012) Cannabidiol protects oligodendrocyte progenitor cells from inflammation-induced apoptosis by attenuating endoplasmic reticulum stress. Cell Death Dis 3:e331PubMedPubMedCentralCrossRef
276.
go back to reference Thomas A, Baillie GL, Phillips AM, Razdan RK, Ross RA, Pertwee RG (2007) Cannabidiol displays unexpectedly high potency as an antagonist of CB1 and CB2 receptor agonists in vitro. Br J Pharmacol 150:613–623PubMedPubMedCentralCrossRef Thomas A, Baillie GL, Phillips AM, Razdan RK, Ross RA, Pertwee RG (2007) Cannabidiol displays unexpectedly high potency as an antagonist of CB1 and CB2 receptor agonists in vitro. Br J Pharmacol 150:613–623PubMedPubMedCentralCrossRef
277.
go back to reference Comelli F, Bettoni I, Colleoni M, Giagnoni G, Costa B (2009) Beneficial effects of a cannabis sativa extract treatment on diabetes-induced neuropathy and oxidative stress. Phytother Res 23:1678–1684PubMedCrossRef Comelli F, Bettoni I, Colleoni M, Giagnoni G, Costa B (2009) Beneficial effects of a cannabis sativa extract treatment on diabetes-induced neuropathy and oxidative stress. Phytother Res 23:1678–1684PubMedCrossRef
278.
go back to reference Comelli F, Giagnoni G, Bettoni I, Colleoni M, Costa B (2008) Antihyperalgesic effect of a cannabis sativa extract in a rat model of neuropathic pain: mechanisms involved. Phytother Res 22:1017–1024PubMedCrossRef Comelli F, Giagnoni G, Bettoni I, Colleoni M, Costa B (2008) Antihyperalgesic effect of a cannabis sativa extract in a rat model of neuropathic pain: mechanisms involved. Phytother Res 22:1017–1024PubMedCrossRef
279.
go back to reference Kohli DR, Li Y, Khasabov SG et al (2010) Pain-related behaviors and neurochemical alterations in mice expressing sickle hemoglobin: modulation by cannabinoids. Blood 116:456–465PubMedPubMedCentralCrossRef Kohli DR, Li Y, Khasabov SG et al (2010) Pain-related behaviors and neurochemical alterations in mice expressing sickle hemoglobin: modulation by cannabinoids. Blood 116:456–465PubMedPubMedCentralCrossRef
280.
go back to reference Howard J, Anie KA, Holdcroft A, Korn S, Davies SC (2005) Cannabis use in sickle cell disease: a questionnaire study. Br J Haematol 131:123–128PubMedCrossRef Howard J, Anie KA, Holdcroft A, Korn S, Davies SC (2005) Cannabis use in sickle cell disease: a questionnaire study. Br J Haematol 131:123–128PubMedCrossRef
281.
go back to reference Maione S, Piscitelli F, Gatta L et al (2011) Non-psychoactive cannabinoids modulate the descending pathway of antinociception in anaesthetized rats through several mechanisms of action. Br J Pharmacol 162:584–596PubMedPubMedCentralCrossRef Maione S, Piscitelli F, Gatta L et al (2011) Non-psychoactive cannabinoids modulate the descending pathway of antinociception in anaesthetized rats through several mechanisms of action. Br J Pharmacol 162:584–596PubMedPubMedCentralCrossRef
282.
283.
go back to reference Russo E, Guy GW (2006) A tale of two cannabinoids: the therapeutic rationale for combining tetrahydrocannabinol and cannabidiol. Med Hypotheses 66:234–246PubMedCrossRef Russo E, Guy GW (2006) A tale of two cannabinoids: the therapeutic rationale for combining tetrahydrocannabinol and cannabidiol. Med Hypotheses 66:234–246PubMedCrossRef
284.
go back to reference Murillo-Rodriguez E, Millan-Aldaco D, Palomero-Rivero M, Mechoulam R, Drucker-Colin R (2006) Cannabidiol, a constituent of cannabis sativa, modulates sleep in rats. FEBS Lett 580:4337–4345PubMedCrossRef Murillo-Rodriguez E, Millan-Aldaco D, Palomero-Rivero M, Mechoulam R, Drucker-Colin R (2006) Cannabidiol, a constituent of cannabis sativa, modulates sleep in rats. FEBS Lett 580:4337–4345PubMedCrossRef
285.
go back to reference Nicholson AN, Turner C, Stone BM, Robson PJ (2004) Effect of Delta-9-tetrahydrocannabinol and cannabidiol on nocturnal sleep and early-morning behavior in young adults. J Clin Psychopharmacol 24:305–313PubMedCrossRef Nicholson AN, Turner C, Stone BM, Robson PJ (2004) Effect of Delta-9-tetrahydrocannabinol and cannabidiol on nocturnal sleep and early-morning behavior in young adults. J Clin Psychopharmacol 24:305–313PubMedCrossRef
286.
go back to reference Zuardi AW, Hallak JE, Crippa JA (2012) Interaction between cannabidiol (CBD) and (9)-tetrahydrocannabinol (THC): influence of administration interval and dose ratio between the cannabinoids. Psychopharmacology 219:247–249PubMedCrossRef Zuardi AW, Hallak JE, Crippa JA (2012) Interaction between cannabidiol (CBD) and (9)-tetrahydrocannabinol (THC): influence of administration interval and dose ratio between the cannabinoids. Psychopharmacology 219:247–249PubMedCrossRef
287.
go back to reference Zuardi AW, Shirakawa I, Finkelfarb E, Karniol IG (1982) Action of cannabidiol on the anxiety and other effects produced by delta 9-THC in normal subjects. Psychopharmacology 76:245–250PubMedCrossRef Zuardi AW, Shirakawa I, Finkelfarb E, Karniol IG (1982) Action of cannabidiol on the anxiety and other effects produced by delta 9-THC in normal subjects. Psychopharmacology 76:245–250PubMedCrossRef
288.
go back to reference Zuardi AW, Finkelfarb E, Bueno OF, Musty RE, Karniol IG (1981) Characteristics of the stimulus produced by the mixture of cannabidiol with delta 9-tetrahydrocannabinol. Arch Int Pharmacodyn Ther 249:137–146PubMed Zuardi AW, Finkelfarb E, Bueno OF, Musty RE, Karniol IG (1981) Characteristics of the stimulus produced by the mixture of cannabidiol with delta 9-tetrahydrocannabinol. Arch Int Pharmacodyn Ther 249:137–146PubMed
289.
go back to reference Bisogno T, Hanus L, De Petrocellis L et al (2001) 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 134:845–852PubMedPubMedCentralCrossRef Bisogno T, Hanus L, De Petrocellis L et al (2001) 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 134:845–852PubMedPubMedCentralCrossRef
290.
go back to reference Ligresti A, Moriello AS, Starowicz K et al (2006) Antitumor activity of plant cannabinoids with emphasis on the effect of cannabidiol on human breast carcinoma. J Pharmacol Exp Ther 318:1375–1387PubMedCrossRef Ligresti A, Moriello AS, Starowicz K et al (2006) Antitumor activity of plant cannabinoids with emphasis on the effect of cannabidiol on human breast carcinoma. J Pharmacol Exp Ther 318:1375–1387PubMedCrossRef
291.
go back to reference Ahrens J, Demir R, Leuwer M et al (2009) The nonpsychotropic cannabinoid cannabidiol modulates and directly activates alpha-1 and alpha-1-Beta glycine receptor function. Pharmacology 83:217–222PubMedCrossRef Ahrens J, Demir R, Leuwer M et al (2009) The nonpsychotropic cannabinoid cannabidiol modulates and directly activates alpha-1 and alpha-1-Beta glycine receptor function. Pharmacology 83:217–222PubMedCrossRef
292.
go back to reference Qin N, Neeper MP, Liu Y, Hutchinson TL, Lubin ML, Flores CM (2008) TRPV2 is activated by cannabidiol and mediates CGRP release in cultured rat dorsal root ganglion neurons. J Neurosci 28:6231–6238PubMedCrossRef Qin N, Neeper MP, Liu Y, Hutchinson TL, Lubin ML, Flores CM (2008) TRPV2 is activated by cannabidiol and mediates CGRP release in cultured rat dorsal root ganglion neurons. J Neurosci 28:6231–6238PubMedCrossRef
293.
go back to reference Ross HR, Napier I, Connor M (2008) Inhibition of recombinant human T-type calcium channels by Delta9-tetrahydrocannabinol and cannabidiol. J Biol Chem 283:16124–16134PubMedPubMedCentralCrossRef Ross HR, Napier I, Connor M (2008) Inhibition of recombinant human T-type calcium channels by Delta9-tetrahydrocannabinol and cannabidiol. J Biol Chem 283:16124–16134PubMedPubMedCentralCrossRef
294.
go back to reference Jenny M, Santer E, Pirich E, Schennach H, Fuchs D (2009) Delta9-tetrahydrocannabinol and cannabidiol modulate mitogen-induced tryptophan degradation and neopterin formation in peripheral blood mononuclear cells in vitro. J Neuroimmunol 207:75–82PubMedCrossRef Jenny M, Santer E, Pirich E, Schennach H, Fuchs D (2009) Delta9-tetrahydrocannabinol and cannabidiol modulate mitogen-induced tryptophan degradation and neopterin formation in peripheral blood mononuclear cells in vitro. J Neuroimmunol 207:75–82PubMedCrossRef
295.
go back to reference Evans AT, Formukong E, Evans FJ (1987) Activation of phospholipase A2 by cannabinoids. Lack of correlation with CNS effects. FEBS Lett 211:119–122PubMedCrossRef Evans AT, Formukong E, Evans FJ (1987) Activation of phospholipase A2 by cannabinoids. Lack of correlation with CNS effects. FEBS Lett 211:119–122PubMedCrossRef
296.
go back to reference Russo EB, Burnett A, Hall B, Parker KK (2005) Agonistic properties of cannabidiol at 5-HT1a receptors. Neurochem Res 30:1037–1043PubMedCrossRef Russo EB, Burnett A, Hall B, Parker KK (2005) Agonistic properties of cannabidiol at 5-HT1a receptors. Neurochem Res 30:1037–1043PubMedCrossRef
297.
go back to reference Drysdale AJ, Ryan D, Pertwee RG, Platt B (2006) Cannabidiol-induced intracellular Ca2+ elevations in hippocampal cells. Neuropharmacology 50:621–631PubMedCrossRef Drysdale AJ, Ryan D, Pertwee RG, Platt B (2006) Cannabidiol-induced intracellular Ca2+ elevations in hippocampal cells. Neuropharmacology 50:621–631PubMedCrossRef
298.
go back to reference Ryan D, Drysdale AJ, Lafourcade C, Pertwee RG, Platt B (2009) Cannabidiol targets mitochondria to regulate intracellular Ca2+ levels. J Neurosci 29:2053–2063PubMedCrossRef Ryan D, Drysdale AJ, Lafourcade C, Pertwee RG, Platt B (2009) Cannabidiol targets mitochondria to regulate intracellular Ca2+ levels. J Neurosci 29:2053–2063PubMedCrossRef
299.
go back to reference Carrier EJ, Auchampach JA, Hillard CJ (2006) Inhibition of an equilibrative nucleoside transporter by cannabidiol: a mechanism of cannabinoid immunosuppression. Proc Natl Acad Sci U S A 103:7895–7900PubMedPubMedCentralCrossRef Carrier EJ, Auchampach JA, Hillard CJ (2006) Inhibition of an equilibrative nucleoside transporter by cannabidiol: a mechanism of cannabinoid immunosuppression. Proc Natl Acad Sci U S A 103:7895–7900PubMedPubMedCentralCrossRef
300.
go back to reference O'Sullivan SE, Sun Y, Bennett AJ, Randall MD, Kendall DA (2009) Time-dependent vascular actions of cannabidiol in the rat aorta. Eur J Pharmacol 612:61–68PubMedCrossRef O'Sullivan SE, Sun Y, Bennett AJ, Randall MD, Kendall DA (2009) Time-dependent vascular actions of cannabidiol in the rat aorta. Eur J Pharmacol 612:61–68PubMedCrossRef
301.
go back to reference Takeda S, Usami N, Yamamoto I, Watanabe K (2009) Cannabidiol-2′,6′-dimethyl ether, a cannabidiol derivative, is a highly potent and selective 15-lipoxygenase inhibitor. Drug Metab Dispos 37:1733–1737PubMedCrossRef Takeda S, Usami N, Yamamoto I, Watanabe K (2009) Cannabidiol-2′,6′-dimethyl ether, a cannabidiol derivative, is a highly potent and selective 15-lipoxygenase inhibitor. Drug Metab Dispos 37:1733–1737PubMedCrossRef
302.
go back to reference Walter L, Franklin A, Witting A et al (2003) Nonpsychotropic cannabinoid receptors regulate microglial cell migration. J Neurosci 23:1398–1405PubMedCrossRef Walter L, Franklin A, Witting A et al (2003) Nonpsychotropic cannabinoid receptors regulate microglial cell migration. J Neurosci 23:1398–1405PubMedCrossRef
303.
go back to reference Rock EM, Kopstick RL, Limebeer CL, Parker LA (2013) Tetrahydrocannabinolic acid reduces nausea-induced conditioned gaping in rats and vomiting in Suncus murinus. Br J Pharmacol 170:641–648PubMedPubMedCentralCrossRef Rock EM, Kopstick RL, Limebeer CL, Parker LA (2013) Tetrahydrocannabinolic acid reduces nausea-induced conditioned gaping in rats and vomiting in Suncus murinus. Br J Pharmacol 170:641–648PubMedPubMedCentralCrossRef
304.
go back to reference Takeda S, Misawa K, Yamamoto I, Watanabe K (2008) Cannabidiolic acid as a selective cyclooxygenase-2 inhibitory component in cannabis. Drug Metab Dispos 36:1917–1921PubMedCrossRef Takeda S, Misawa K, Yamamoto I, Watanabe K (2008) Cannabidiolic acid as a selective cyclooxygenase-2 inhibitory component in cannabis. Drug Metab Dispos 36:1917–1921PubMedCrossRef
305.
go back to reference Bolognini D, Rock EM, Cluny NL et al (2013) Cannabidiolic acid prevents vomiting in Suncus murinus and nausea-induced behaviour in rats by enhancing 5-HT1A receptor activation. Br J Pharmacol 168:1456–1470PubMedPubMedCentralCrossRef Bolognini D, Rock EM, Cluny NL et al (2013) Cannabidiolic acid prevents vomiting in Suncus murinus and nausea-induced behaviour in rats by enhancing 5-HT1A receptor activation. Br J Pharmacol 168:1456–1470PubMedPubMedCentralCrossRef
306.
go back to reference Buchbauer G, Bohusch R (2015) Biological activities of essential oils: an update. In: Husnu Can Baser K, Buchbauer G (eds) Handbook of essential oils: science, technology, and applications, 2nd edn. CRC Press, Boca Raton, pp 281–322CrossRef Buchbauer G, Bohusch R (2015) Biological activities of essential oils: an update. In: Husnu Can Baser K, Buchbauer G (eds) Handbook of essential oils: science, technology, and applications, 2nd edn. CRC Press, Boca Raton, pp 281–322CrossRef
307.
go back to reference Bowles EJ (2004) The chemistry of aromatherapeutic oils. Allen & Unwin, Crows Nest Bowles EJ (2004) The chemistry of aromatherapeutic oils. Allen & Unwin, Crows Nest
308.
go back to reference Paduch R, Kandefer-Szerszen M, Trytek M, Fiedurek J (2007) Terpenes: substances useful in human healthcare. Arch Immunol Ther Exp 55:315–327CrossRef Paduch R, Kandefer-Szerszen M, Trytek M, Fiedurek J (2007) Terpenes: substances useful in human healthcare. Arch Immunol Ther Exp 55:315–327CrossRef
309.
go back to reference Noma Y, Asakawa Y (2010) Biotransformation of monoterpenoids by microorganisms, insects, and mammals. In: Baser KHC, Buchbauer G (eds) Handbook of essential oils: science, technology, and applications. CRC Press, Boca Raton, pp 585–736 Noma Y, Asakawa Y (2010) Biotransformation of monoterpenoids by microorganisms, insects, and mammals. In: Baser KHC, Buchbauer G (eds) Handbook of essential oils: science, technology, and applications. CRC Press, Boca Raton, pp 585–736
310.
go back to reference Rufino AT, Ribeiro M, Judas F et al (2014) Anti-inflammatory and chondroprotective activity of (+)-alpha-pinene: structural and enantiomeric selectivity. J Nat Prod 77:264–269PubMedCrossRef Rufino AT, Ribeiro M, Judas F et al (2014) Anti-inflammatory and chondroprotective activity of (+)-alpha-pinene: structural and enantiomeric selectivity. J Nat Prod 77:264–269PubMedCrossRef
311.
go back to reference Neves A, Rosa S, Goncalves J et al (2010) Screening of five essential oils for identification of potential inhibitors of IL-1-induced Nf-kappaB activation and NO production in human chondrocytes: characterization of the inhibitory activity of alpha-pinene. Planta Med 76:303–308PubMedCrossRef Neves A, Rosa S, Goncalves J et al (2010) Screening of five essential oils for identification of potential inhibitors of IL-1-induced Nf-kappaB activation and NO production in human chondrocytes: characterization of the inhibitory activity of alpha-pinene. Planta Med 76:303–308PubMedCrossRef
312.
go back to reference Gil ML, Jimenez J, Ocete MA, Zarzuelo A, Cabo MM (1989) Comparative study of different essential oils of bupleurum gibraltaricum lamarck. Pharmazie 44:284–287PubMed Gil ML, Jimenez J, Ocete MA, Zarzuelo A, Cabo MM (1989) Comparative study of different essential oils of bupleurum gibraltaricum lamarck. Pharmazie 44:284–287PubMed
313.
go back to reference Him A, Ozbek H, Turel I, Oner AC (2008) Antinociceptive activity of α-pinene and fenchone. Pharmacol Online 3:363–369 Him A, Ozbek H, Turel I, Oner AC (2008) Antinociceptive activity of α-pinene and fenchone. Pharmacol Online 3:363–369
314.
go back to reference Van Cleemput M, Cattoor K, De Bosscher K, Haegeman G, De Keukeleire D, Heyerick A (2009) Hop (Humulus lupulus)-derived bitter acids as multipotent bioactive compounds. J Nat Prod 72:1220–1230PubMedCrossRef Van Cleemput M, Cattoor K, De Bosscher K, Haegeman G, De Keukeleire D, Heyerick A (2009) Hop (Humulus lupulus)-derived bitter acids as multipotent bioactive compounds. J Nat Prod 72:1220–1230PubMedCrossRef
315.
go back to reference Lorenzetti BB, Souza GE, Sarti SJ, Santos Filho D, Ferreira SH (1991) Myrcene mimics the peripheral analgesic activity of lemongrass tea. J Ethnopharmacol 34:43–48PubMedCrossRef Lorenzetti BB, Souza GE, Sarti SJ, Santos Filho D, Ferreira SH (1991) Myrcene mimics the peripheral analgesic activity of lemongrass tea. J Ethnopharmacol 34:43–48PubMedCrossRef
316.
go back to reference Rao VS, Menezes AM, Viana GS (1990) Effect of myrcene on nociception in mice. J Pharm Pharmacol 42:877–878PubMedCrossRef Rao VS, Menezes AM, Viana GS (1990) Effect of myrcene on nociception in mice. J Pharm Pharmacol 42:877–878PubMedCrossRef
317.
go back to reference do Vale TG, Furtado EC, Santos JG Jr, Viana GS (2002) Central effects of citral, myrcene and limonene, constituents of essential oil chemotypes from lippia alba (Mill.) n.e. brown. Phytomedicine 9:709–714PubMedCrossRef do Vale TG, Furtado EC, Santos JG Jr, Viana GS (2002) Central effects of citral, myrcene and limonene, constituents of essential oil chemotypes from lippia alba (Mill.) n.e. brown. Phytomedicine 9:709–714PubMedCrossRef
318.
go back to reference Bisset NG, Wichtl M (2004) Herbal drugs and phytopharmaceuticals: a handbook for practice on a scientific basis, 3rd edn. Medpharm Scientific Publishers: Stuttgart; CRC Press, Boca Raton Bisset NG, Wichtl M (2004) Herbal drugs and phytopharmaceuticals: a handbook for practice on a scientific basis, 3rd edn. Medpharm Scientific Publishers: Stuttgart; CRC Press, Boca Raton
319.
go back to reference Souza MC, Siani AC, Ramos MF, Menezes-de-Lima OJ, Henriques MG (2003) Evaluation of anti-inflammatory activity of essential oils from two Asteraceae species. Pharmazie 58:582–586PubMed Souza MC, Siani AC, Ramos MF, Menezes-de-Lima OJ, Henriques MG (2003) Evaluation of anti-inflammatory activity of essential oils from two Asteraceae species. Pharmazie 58:582–586PubMed
320.
go back to reference Rufino AT, Ribeiro M, Sousa C et al (2015) Evaluation of the anti-inflammatory, anti-catabolic and pro-anabolic effects of E-caryophyllene, myrcene and limonene in a cell model of osteoarthritis. Eur J Pharmacol 750:141–150PubMedCrossRef Rufino AT, Ribeiro M, Sousa C et al (2015) Evaluation of the anti-inflammatory, anti-catabolic and pro-anabolic effects of E-caryophyllene, myrcene and limonene in a cell model of osteoarthritis. Eur J Pharmacol 750:141–150PubMedCrossRef
321.
go back to reference Piccinelli AC, Santos JA, Konkiewitz EC et al (2015) Antihyperalgesic and antidepressive actions of (R)-(+)-limonene, alpha-phellandrene, and essential oil from Schinus terebinthifolius fruits in a neuropathic pain model. Nutr Neurosci 18:217–224PubMedCrossRef Piccinelli AC, Santos JA, Konkiewitz EC et al (2015) Antihyperalgesic and antidepressive actions of (R)-(+)-limonene, alpha-phellandrene, and essential oil from Schinus terebinthifolius fruits in a neuropathic pain model. Nutr Neurosci 18:217–224PubMedCrossRef
322.
go back to reference Hirota R, Roger NN, Nakamura H, Song HS, Sawamura M, Suganuma N (2010) Anti-inflammatory effects of limonene from yuzu (Citrus junos Tanaka) essential oil on eosinophils. J Food Sci 75:H87–H92PubMedCrossRef Hirota R, Roger NN, Nakamura H, Song HS, Sawamura M, Suganuma N (2010) Anti-inflammatory effects of limonene from yuzu (Citrus junos Tanaka) essential oil on eosinophils. J Food Sci 75:H87–H92PubMedCrossRef
323.
go back to reference Chaudhary SC, Siddiqui MS, Athar M, Alam MS (2012) D-limonene modulates inflammation, oxidative stress and Ras-ERK pathway to inhibit murine skin tumorigenesis. Hum Exp Toxicol 31:798–811PubMedCrossRef Chaudhary SC, Siddiqui MS, Athar M, Alam MS (2012) D-limonene modulates inflammation, oxidative stress and Ras-ERK pathway to inhibit murine skin tumorigenesis. Hum Exp Toxicol 31:798–811PubMedCrossRef
324.
go back to reference d'Alessio PA, Ostan R, Bisson JF, Schulzke JD, Ursini MV, Bene MC (2013) Oral administration of d-limonene controls inflammation in rat colitis and displays anti-inflammatory properties as diet supplementation in humans. Life Sci 92:1151–1156PubMedCrossRef d'Alessio PA, Ostan R, Bisson JF, Schulzke JD, Ursini MV, Bene MC (2013) Oral administration of d-limonene controls inflammation in rat colitis and displays anti-inflammatory properties as diet supplementation in humans. Life Sci 92:1151–1156PubMedCrossRef
325.
go back to reference Kim MJ, Yang KW, Kim SS et al (2014) Chemical composition and anti-inflammation activity of essential oils from citrus unshiu flower. Nat Prod Commun 9:727–730PubMed Kim MJ, Yang KW, Kim SS et al (2014) Chemical composition and anti-inflammation activity of essential oils from citrus unshiu flower. Nat Prod Commun 9:727–730PubMed
326.
go back to reference Komori T, Fujiwara R, Tanida M, Nomura J, Yokoyama MM (1995) Effects of citrus fragrance on immune function and depressive states. Neuroimmunomodulation 2:174–180PubMedCrossRef Komori T, Fujiwara R, Tanida M, Nomura J, Yokoyama MM (1995) Effects of citrus fragrance on immune function and depressive states. Neuroimmunomodulation 2:174–180PubMedCrossRef
327.
go back to reference de Almeida AA, Costa JP, de Carvalho RB, de Sousa DP, de Freitas RM (2012) Evaluation of acute toxicity of a natural compound (+)-limonene epoxide and its anxiolytic-like action. Brain Res 1448:56–62PubMedCrossRef de Almeida AA, Costa JP, de Carvalho RB, de Sousa DP, de Freitas RM (2012) Evaluation of acute toxicity of a natural compound (+)-limonene epoxide and its anxiolytic-like action. Brain Res 1448:56–62PubMedCrossRef
328.
go back to reference Carvalho-Freitas MI, Costa M (2002) Anxiolytic and sedative effects of extracts and essential oil from citrus aurantium L. Biol Pharm Bull 25:1629–1633PubMedCrossRef Carvalho-Freitas MI, Costa M (2002) Anxiolytic and sedative effects of extracts and essential oil from citrus aurantium L. Biol Pharm Bull 25:1629–1633PubMedCrossRef
329.
go back to reference Pultrini Ade M, Galindo LA, Costa M (2006) Effects of the essential oil from citrus aurantium L. in experimental anxiety models in mice. Life Sci 78:1720–1725PubMedCrossRef Pultrini Ade M, Galindo LA, Costa M (2006) Effects of the essential oil from citrus aurantium L. in experimental anxiety models in mice. Life Sci 78:1720–1725PubMedCrossRef
330.
go back to reference Saiyudthong S, Marsden CA (2011) Acute effects of bergamot oil on anxiety-related behaviour and corticosterone level in rats. Phytother Res 25:858–862PubMedCrossRef Saiyudthong S, Marsden CA (2011) Acute effects of bergamot oil on anxiety-related behaviour and corticosterone level in rats. Phytother Res 25:858–862PubMedCrossRef
331.
go back to reference Pimenta FC, Alves MF, Pimenta MB et al (2016) Anxiolytic effect of citrus aurantium L. on patients with chronic myeloid leukemia. Phytother Res 30:613–617PubMedCrossRef Pimenta FC, Alves MF, Pimenta MB et al (2016) Anxiolytic effect of citrus aurantium L. on patients with chronic myeloid leukemia. Phytother Res 30:613–617PubMedCrossRef
332.
go back to reference Komiya M, Takeuchi T, Harada E (2006) Lemon oil vapor causes an anti-stress effect via modulating the 5-HT and DA activities in mice. Behav Brain Res 172:240–249PubMedCrossRef Komiya M, Takeuchi T, Harada E (2006) Lemon oil vapor causes an anti-stress effect via modulating the 5-HT and DA activities in mice. Behav Brain Res 172:240–249PubMedCrossRef
333.
go back to reference Peana AT, D'Aquila PS, Chessa ML, Moretti MD, Serra G, Pippia P (2003) (−)-linalool produces antinociception in two experimental models of pain. Eur J Pharmacol 460:37–41PubMedCrossRef Peana AT, D'Aquila PS, Chessa ML, Moretti MD, Serra G, Pippia P (2003) (−)-linalool produces antinociception in two experimental models of pain. Eur J Pharmacol 460:37–41PubMedCrossRef
334.
go back to reference Peana AT, D'Aquila PS, Panin F, Serra G, Pippia P, Moretti MD (2002) Anti-inflammatory activity of linalool and linalyl acetate constituents of essential oils. Phytomedicine 9:721–726PubMedCrossRef Peana AT, D'Aquila PS, Panin F, Serra G, Pippia P, Moretti MD (2002) Anti-inflammatory activity of linalool and linalyl acetate constituents of essential oils. Phytomedicine 9:721–726PubMedCrossRef
335.
go back to reference Peana AT, Marzocco S, Popolo A, Pinto A (2006) (−)-linalool inhibits in vitro NO formation: probable involvement in the antinociceptive activity of this monoterpene compound. Life Sci 78:719–723PubMedCrossRef Peana AT, Marzocco S, Popolo A, Pinto A (2006) (−)-linalool inhibits in vitro NO formation: probable involvement in the antinociceptive activity of this monoterpene compound. Life Sci 78:719–723PubMedCrossRef
336.
go back to reference de Sousa DP, Nobrega FF, Santos CC, de Almeida RN (2010) Anticonvulsant activity of the linalool enantiomers and racemate: investigation of chiral influence. Nat Prod Commun 5:1847–1851PubMed de Sousa DP, Nobrega FF, Santos CC, de Almeida RN (2010) Anticonvulsant activity of the linalool enantiomers and racemate: investigation of chiral influence. Nat Prod Commun 5:1847–1851PubMed
337.
go back to reference Elisabetsky E, Marschner J, Souza DO (1995) Effects of linalool on glutamatergic system in the rat cerebral cortex. Neurochem Res 20:461–465PubMedCrossRef Elisabetsky E, Marschner J, Souza DO (1995) Effects of linalool on glutamatergic system in the rat cerebral cortex. Neurochem Res 20:461–465PubMedCrossRef
338.
go back to reference Ismail M (2006) Central properties and chemical composition of Ocimum basilicum essential oil. Pharm Biol 44:619–626CrossRef Ismail M (2006) Central properties and chemical composition of Ocimum basilicum essential oil. Pharm Biol 44:619–626CrossRef
339.
go back to reference Silva Brum LF, Emanuelli T, Souza DO, Elisabetsky E (2001) Effects of linalool on glutamate release and uptake in mouse cortical synaptosomes. Neurochem Res 26:191–194PubMedCrossRef Silva Brum LF, Emanuelli T, Souza DO, Elisabetsky E (2001) Effects of linalool on glutamate release and uptake in mouse cortical synaptosomes. Neurochem Res 26:191–194PubMedCrossRef
340.
go back to reference Nunes DS, Linck VM, da Silva AL, Figueiro M, Elisabetsky E (2010) Psychopharmacology of essential oils. In: Baser KHC, Buchbauer G (eds) Handbook of essential oils: science, technology, and applications. CRC Press, Boca Raton, pp 297–314 Nunes DS, Linck VM, da Silva AL, Figueiro M, Elisabetsky E (2010) Psychopharmacology of essential oils. In: Baser KHC, Buchbauer G (eds) Handbook of essential oils: science, technology, and applications. CRC Press, Boca Raton, pp 297–314
341.
go back to reference Nakamura A, Fujiwara S, Matsumoto I, Abe K (2009) Stress repression in restrained rats by (R)-(−)-linalool inhalation and gene expression profiling of their whole blood cells. J Agric Food Chem 57:5480–5485PubMedCrossRef Nakamura A, Fujiwara S, Matsumoto I, Abe K (2009) Stress repression in restrained rats by (R)-(−)-linalool inhalation and gene expression profiling of their whole blood cells. J Agric Food Chem 57:5480–5485PubMedCrossRef
342.
go back to reference Russo EB (2001) Handbook of psychotropic herbs: a scientific analysis of herbal remedies for psychiatric conditions. Haworth Press, Binghamton Russo EB (2001) Handbook of psychotropic herbs: a scientific analysis of herbal remedies for psychiatric conditions. Haworth Press, Binghamton
343.
go back to reference Cline M, Taylor JE, Flores J, Bracken S, McCall S, Ceremuga TE (2008) Investigation of the anxiolytic effects of linalool, a lavender extract, in the male Sprague-Dawley rat. AANA J 76:47–52PubMed Cline M, Taylor JE, Flores J, Bracken S, McCall S, Ceremuga TE (2008) Investigation of the anxiolytic effects of linalool, a lavender extract, in the male Sprague-Dawley rat. AANA J 76:47–52PubMed
344.
go back to reference Cheng BH, Sheen LY, Chang ST (2014) Evaluation of anxiolytic potency of essential oil and S-(+)-linalool from Cinnamomum osmophloeum ct. linalool leaves in mice. J Tradit Complement Med 5:27–34PubMedPubMedCentralCrossRef Cheng BH, Sheen LY, Chang ST (2014) Evaluation of anxiolytic potency of essential oil and S-(+)-linalool from Cinnamomum osmophloeum ct. linalool leaves in mice. J Tradit Complement Med 5:27–34PubMedPubMedCentralCrossRef
345.
go back to reference Buchbauer G, Jirovetz L, Jager W, Dietrich H, Plank C (1991) Aromatherapy: evidence for sedative effects of the essential oil of lavender after inhalation. Z Naturforsch C 46:1067–1072PubMedCrossRef Buchbauer G, Jirovetz L, Jager W, Dietrich H, Plank C (1991) Aromatherapy: evidence for sedative effects of the essential oil of lavender after inhalation. Z Naturforsch C 46:1067–1072PubMedCrossRef
346.
go back to reference Jirovetz L, Buchbauer G, Jager W, Woidich A, Nikiforov A (1992) Analysis of fragrance compounds in blood samples of mice by gas chromatography, mass spectrometry, GC/FTIR and GC/AES after inhalation of sandalwood oil. Biomed Chromatogr 6:133–134PubMedCrossRef Jirovetz L, Buchbauer G, Jager W, Woidich A, Nikiforov A (1992) Analysis of fragrance compounds in blood samples of mice by gas chromatography, mass spectrometry, GC/FTIR and GC/AES after inhalation of sandalwood oil. Biomed Chromatogr 6:133–134PubMedCrossRef
347.
go back to reference Buchbauer G, Jirovetz L, Jager W, Plank C, Dietrich H (1993) Fragrance compounds and essential oils with sedative effects upon inhalation. J Pharm Sci 82:660–664PubMedCrossRef Buchbauer G, Jirovetz L, Jager W, Plank C, Dietrich H (1993) Fragrance compounds and essential oils with sedative effects upon inhalation. J Pharm Sci 82:660–664PubMedCrossRef
348.
go back to reference Re L, Barocci S, Sonnino S et al (2000) Linalool modifies the nicotinic receptor-ion channel kinetics at the mouse neuromuscular junction. Pharmacol Res 42:177–182PubMedCrossRef Re L, Barocci S, Sonnino S et al (2000) Linalool modifies the nicotinic receptor-ion channel kinetics at the mouse neuromuscular junction. Pharmacol Res 42:177–182PubMedCrossRef
349.
go back to reference Ghelardini C, Galeotti N, Salvatore G, Mazzanti G (1999) Local anaesthetic activity of the essential oil of lavandula angustifolia. Planta Med 65:700–703PubMedCrossRef Ghelardini C, Galeotti N, Salvatore G, Mazzanti G (1999) Local anaesthetic activity of the essential oil of lavandula angustifolia. Planta Med 65:700–703PubMedCrossRef
350.
go back to reference Peana AT, Rubattu P, Piga GG et al (2006) Involvement of adenosine A1 and A2A receptors in (−)-linalool-induced antinociception. Life Sci 78:2471–2474PubMedCrossRef Peana AT, Rubattu P, Piga GG et al (2006) Involvement of adenosine A1 and A2A receptors in (−)-linalool-induced antinociception. Life Sci 78:2471–2474PubMedCrossRef
351.
go back to reference Batista PA, Werner MF, Oliveira EC et al (2008) Evidence for the involvement of ionotropic glutamatergic receptors on the antinociceptive effect of (−)-linalool in mice. Neurosci Lett 440:299–303PubMedCrossRef Batista PA, Werner MF, Oliveira EC et al (2008) Evidence for the involvement of ionotropic glutamatergic receptors on the antinociceptive effect of (−)-linalool in mice. Neurosci Lett 440:299–303PubMedCrossRef
352.
go back to reference Kim JT, Ren CJ, Fielding GA et al (2007) Treatment with lavender aromatherapy in the post-anesthesia care unit reduces opioid requirements of morbidly obese patients undergoing laparoscopic adjustable gastric banding. Obes Surg 17:920–925PubMedCrossRef Kim JT, Ren CJ, Fielding GA et al (2007) Treatment with lavender aromatherapy in the post-anesthesia care unit reduces opioid requirements of morbidly obese patients undergoing laparoscopic adjustable gastric banding. Obes Surg 17:920–925PubMedCrossRef
353.
go back to reference Klauke AL, Racz I, Pradier B et al (2014) The cannabinoid CB(2) receptor-selective phytocannabinoid beta-caryophyllene exerts analgesic effects in mouse models of inflammatory and neuropathic pain. Eur Neuropsychopharmacol 24:608–620PubMedCrossRef Klauke AL, Racz I, Pradier B et al (2014) The cannabinoid CB(2) receptor-selective phytocannabinoid beta-caryophyllene exerts analgesic effects in mouse models of inflammatory and neuropathic pain. Eur Neuropsychopharmacol 24:608–620PubMedCrossRef
354.
go back to reference Passos GF, Fernandes ES, da Cunha FM et al (2007) Anti-inflammatory and anti-allergic properties of the essential oil and active compounds from Cordia verbenacea. J Ethnopharmacol 110:323–333PubMedCrossRef Passos GF, Fernandes ES, da Cunha FM et al (2007) Anti-inflammatory and anti-allergic properties of the essential oil and active compounds from Cordia verbenacea. J Ethnopharmacol 110:323–333PubMedCrossRef
355.
go back to reference Rogerio AP, Andrade EL, Leite DF, Figueiredo CP, Calixto JB (2009) Preventive and therapeutic anti-inflammatory properties of the sesquiterpene alpha-humulene in experimental airways allergic inflammation. Br J Pharmacol 158:1074–1087PubMedPubMedCentralCrossRef Rogerio AP, Andrade EL, Leite DF, Figueiredo CP, Calixto JB (2009) Preventive and therapeutic anti-inflammatory properties of the sesquiterpene alpha-humulene in experimental airways allergic inflammation. Br J Pharmacol 158:1074–1087PubMedPubMedCentralCrossRef
356.
go back to reference Medeiros R, Passos GF, Vitor CE et al (2007) Effect of two active compounds obtained from the essential oil of cordia verbenacea on the acute inflammatory responses elicited by LPS in the rat paw. Br J Pharmacol 151:618–627PubMedPubMedCentralCrossRef Medeiros R, Passos GF, Vitor CE et al (2007) Effect of two active compounds obtained from the essential oil of cordia verbenacea on the acute inflammatory responses elicited by LPS in the rat paw. Br J Pharmacol 151:618–627PubMedPubMedCentralCrossRef
357.
go back to reference Horvath B, Mukhopadhyay P, Kechrid M et al (2012) beta-caryophyllene ameliorates cisplatin-induced nephrotoxicity in a cannabinoid 2 receptor-dependent manner. Free Radic Biol Med 52:1325–1333PubMedPubMedCentralCrossRef Horvath B, Mukhopadhyay P, Kechrid M et al (2012) beta-caryophyllene ameliorates cisplatin-induced nephrotoxicity in a cannabinoid 2 receptor-dependent manner. Free Radic Biol Med 52:1325–1333PubMedPubMedCentralCrossRef
358.
go back to reference Ghelardini C, Galeotti N, Di Cesare Mannelli L, Mazzanti G, Bartolini A (2001) Local anaesthetic activity of beta-caryophyllene. Farmaco 56:387–389PubMedCrossRef Ghelardini C, Galeotti N, Di Cesare Mannelli L, Mazzanti G, Bartolini A (2001) Local anaesthetic activity of beta-caryophyllene. Farmaco 56:387–389PubMedCrossRef
359.
go back to reference Basile AC, Sertie JA, Freitas PC, Zanini AC (1988) Anti-inflammatory activity of oleoresin from Brazilian Copaifera. J Ethnopharmacol 22:101–109PubMedCrossRef Basile AC, Sertie JA, Freitas PC, Zanini AC (1988) Anti-inflammatory activity of oleoresin from Brazilian Copaifera. J Ethnopharmacol 22:101–109PubMedCrossRef
360.
go back to reference Ozturk A, Ozbek H (2005) The anti-inflammatory activity of Eugenia caryophyllata essential oil: an animal model of anti-inflammatory activity. Eur J Gen Med 2:159–163CrossRef Ozturk A, Ozbek H (2005) The anti-inflammatory activity of Eugenia caryophyllata essential oil: an animal model of anti-inflammatory activity. Eur J Gen Med 2:159–163CrossRef
361.
go back to reference Apel MA, Lima ME, Sobral M et al (2010) Anti-inflammatory activity of essential oil from leaves of Myrciaria tenella and Calycorectes sellowianus. Pharm Biol 48:433–438PubMedCrossRef Apel MA, Lima ME, Sobral M et al (2010) Anti-inflammatory activity of essential oil from leaves of Myrciaria tenella and Calycorectes sellowianus. Pharm Biol 48:433–438PubMedCrossRef
362.
go back to reference Al Mansouri S, Ojha S, Al Maamari E, Al Ameri M, Nurulain SM, Bahi A (2014) The cannabinoid receptor 2 agonist, beta-caryophyllene, reduced voluntary alcohol intake and attenuated ethanol-induced place preference and sensitivity in mice. Pharmacol Biochem Behav 124:260–268PubMedCrossRef Al Mansouri S, Ojha S, Al Maamari E, Al Ameri M, Nurulain SM, Bahi A (2014) The cannabinoid receptor 2 agonist, beta-caryophyllene, reduced voluntary alcohol intake and attenuated ethanol-induced place preference and sensitivity in mice. Pharmacol Biochem Behav 124:260–268PubMedCrossRef
363.
365.
go back to reference Bahi A, Al Mansouri S, Al Memari E, Al Ameri M, Nurulain SM, Ojha S (2014) beta-caryophyllene, a CB2 receptor agonist produces multiple behavioral changes relevant to anxiety and depression in mice. Physiol Behav 135:119–124PubMedCrossRef Bahi A, Al Mansouri S, Al Memari E, Al Ameri M, Nurulain SM, Ojha S (2014) beta-caryophyllene, a CB2 receptor agonist produces multiple behavioral changes relevant to anxiety and depression in mice. Physiol Behav 135:119–124PubMedCrossRef
366.
go back to reference Fernandes ES, Passos GF, Medeiros R et al (2007) Anti-inflammatory effects of compounds alpha-humulene and (−)-trans-caryophyllene isolated from the essential oil of Cordia verbenacea. Eur J Pharmacol 569:228–236PubMedCrossRef Fernandes ES, Passos GF, Medeiros R et al (2007) Anti-inflammatory effects of compounds alpha-humulene and (−)-trans-caryophyllene isolated from the essential oil of Cordia verbenacea. Eur J Pharmacol 569:228–236PubMedCrossRef
367.
go back to reference Chaves JS, Leal PC, Pianowisky L, Calixto JB (2008) Pharmacokinetics and tissue distribution of the sesquiterpene alpha-humulene in mice. Planta Med 74:1678–1683PubMedCrossRef Chaves JS, Leal PC, Pianowisky L, Calixto JB (2008) Pharmacokinetics and tissue distribution of the sesquiterpene alpha-humulene in mice. Planta Med 74:1678–1683PubMedCrossRef
368.
go back to reference Binet L, Binet P, Miocque M, Roux M, Bernier A (1972) Reserches sur les proprietes pharmcodynamiques (action sedative et action spasmolytique) de quelques alcools terpeniques aliphatiques. Ann Pharm Fr 30:611–616PubMed Binet L, Binet P, Miocque M, Roux M, Bernier A (1972) Reserches sur les proprietes pharmcodynamiques (action sedative et action spasmolytique) de quelques alcools terpeniques aliphatiques. Ann Pharm Fr 30:611–616PubMed
369.
go back to reference Maurya AK, Singh M, Dubey V, Srivastava S, Luqman S, Bawankule DU (2014) Alpha-(−)-Bisabolol reduces pro-inflammatory cytokine production and ameliorates skin inflammation. Curr Pharm Biotechnol 15:173–181PubMedCrossRef Maurya AK, Singh M, Dubey V, Srivastava S, Luqman S, Bawankule DU (2014) Alpha-(−)-Bisabolol reduces pro-inflammatory cytokine production and ameliorates skin inflammation. Curr Pharm Biotechnol 15:173–181PubMedCrossRef
370.
go back to reference Nurulain S, Prytkova T, Sultan AM et al (2015) Inhibitory actions of bisabolol on alpha7-nicotinic acetylcholine receptors. Neuroscience 306:91–99PubMedCrossRef Nurulain S, Prytkova T, Sultan AM et al (2015) Inhibitory actions of bisabolol on alpha7-nicotinic acetylcholine receptors. Neuroscience 306:91–99PubMedCrossRef
371.
go back to reference Fischedick JT, Hazekamp A, Erkelens T, Choi YH, Verpoorte R (2010) Metabolic fingerprinting of cannabis sativa L., cannabinoids and terpenoids for chemotaxonomic and drug standardization purposes. Phytochemistry 71:2058–2073PubMedCrossRef Fischedick JT, Hazekamp A, Erkelens T, Choi YH, Verpoorte R (2010) Metabolic fingerprinting of cannabis sativa L., cannabinoids and terpenoids for chemotaxonomic and drug standardization purposes. Phytochemistry 71:2058–2073PubMedCrossRef
372.
go back to reference Hillig KW (2004) A chemotaxonomic analysis of terpenoid variation in Cannabis. Biochem Syst Ecol 32:875–891CrossRef Hillig KW (2004) A chemotaxonomic analysis of terpenoid variation in Cannabis. Biochem Syst Ecol 32:875–891CrossRef
373.
go back to reference Hillig KW, Mahlberg PG (2004) A chemotaxonomic analysis of cannabinoid variation in cannabis (Cannabaceae). Am J Bot 91:966–975PubMedCrossRef Hillig KW, Mahlberg PG (2004) A chemotaxonomic analysis of cannabinoid variation in cannabis (Cannabaceae). Am J Bot 91:966–975PubMedCrossRef
375.
go back to reference Lucas P, Walsh Z (2017) Medical cannabis access, use, and substitution for prescription opioids and other substances: a survey of authorized medical cannabis patients. Int J Drug Policy 42:30–35PubMedCrossRef Lucas P, Walsh Z (2017) Medical cannabis access, use, and substitution for prescription opioids and other substances: a survey of authorized medical cannabis patients. Int J Drug Policy 42:30–35PubMedCrossRef
376.
go back to reference Lucas P, Walsh Z, Crosby K et al (2016) Substituting cannabis for prescription drugs, alcohol and other substances among medical cannabis patients: the impact of contextual factors. Drug Alcohol Rev 35:326–333PubMedCrossRef Lucas P, Walsh Z, Crosby K et al (2016) Substituting cannabis for prescription drugs, alcohol and other substances among medical cannabis patients: the impact of contextual factors. Drug Alcohol Rev 35:326–333PubMedCrossRef
378.
go back to reference Nielsen S, Sabioni P, Trigo JM et al (2017) Opioid-sparing effect of cannabinoids: a systematic review and meta-analysis. Neuropsychopharmacology 42:1752–1765PubMedCrossRefPubMedCentral Nielsen S, Sabioni P, Trigo JM et al (2017) Opioid-sparing effect of cannabinoids: a systematic review and meta-analysis. Neuropsychopharmacology 42:1752–1765PubMedCrossRefPubMedCentral
379.
go back to reference Bushlin I, Rozenfeld R, Devi LA (2010) Cannabinoid-opioid interactions during neuropathic pain and analgesia. Curr Opin Pharmacol 10:80–86PubMedCrossRef Bushlin I, Rozenfeld R, Devi LA (2010) Cannabinoid-opioid interactions during neuropathic pain and analgesia. Curr Opin Pharmacol 10:80–86PubMedCrossRef
380.
go back to reference Parolaro D, Rubino T, Vigano D, Massi P, Guidali C, Realini N (2010) Cellular mechanisms underlying the interaction between cannabinoid and opioid system. Curr Drug Targets 11:393–405PubMedCrossRef Parolaro D, Rubino T, Vigano D, Massi P, Guidali C, Realini N (2010) Cellular mechanisms underlying the interaction between cannabinoid and opioid system. Curr Drug Targets 11:393–405PubMedCrossRef
381.
go back to reference Welch SP, Stevens DL (1992) Antinociceptive activity of intrathecally administered cannabinoids alone, and in combination with morphine, in mice. J Pharmacol Exp Ther 262:10–18PubMed Welch SP, Stevens DL (1992) Antinociceptive activity of intrathecally administered cannabinoids alone, and in combination with morphine, in mice. J Pharmacol Exp Ther 262:10–18PubMed
382.
go back to reference Pugh G Jr, Smith PB, Dombrowski DS, Welch SP (1996) The role of endogenous opioids in enhancing the antinociception produced by the combination of delta 9-tetrahydrocannabinol and morphine in the spinal cord. J Pharmacol Exp Ther 279:608–616PubMed Pugh G Jr, Smith PB, Dombrowski DS, Welch SP (1996) The role of endogenous opioids in enhancing the antinociception produced by the combination of delta 9-tetrahydrocannabinol and morphine in the spinal cord. J Pharmacol Exp Ther 279:608–616PubMed
383.
go back to reference Cichewicz DL, Welch SP, Smith FL (2005) Enhancement of transdermal fentanyl and buprenorphine antinociception by transdermal delta9-tetrahydrocannabinol. Eur J Pharmacol 525:74–82PubMedCrossRef Cichewicz DL, Welch SP, Smith FL (2005) Enhancement of transdermal fentanyl and buprenorphine antinociception by transdermal delta9-tetrahydrocannabinol. Eur J Pharmacol 525:74–82PubMedCrossRef
384.
go back to reference Cichewicz DL, Martin ZL, Smith FL, Welch SP (1999) Enhancement mu opioid antinociception by oral delta9-tetrahydrocannabinol: dose-response analysis and receptor identification. J Pharmacol Exp Ther 289:859–867PubMed Cichewicz DL, Martin ZL, Smith FL, Welch SP (1999) Enhancement mu opioid antinociception by oral delta9-tetrahydrocannabinol: dose-response analysis and receptor identification. J Pharmacol Exp Ther 289:859–867PubMed
386.
go back to reference Haroutounian S, Ratz Y, Ginosar Y et al (2016) The effect of medicinal cannabis on pain and quality-of-life outcomes in chronic pain: a prospective open-label study. Clin J Pain 32:1036–1043PubMedCrossRef Haroutounian S, Ratz Y, Ginosar Y et al (2016) The effect of medicinal cannabis on pain and quality-of-life outcomes in chronic pain: a prospective open-label study. Clin J Pain 32:1036–1043PubMedCrossRef
387.
go back to reference Boehnke KF, Litinas E, Clauw DJ (2016) Medical cannabis use is associated with decreased opiate medication use in a retrospective cross-sectional survey of patients with chronic pain. J Pain 17:739–744PubMedCrossRef Boehnke KF, Litinas E, Clauw DJ (2016) Medical cannabis use is associated with decreased opiate medication use in a retrospective cross-sectional survey of patients with chronic pain. J Pain 17:739–744PubMedCrossRef
388.
go back to reference Livingston MD, Barnett TE, Delcher C, Wagenaar AC (2017) Recreational cannabis legalization and opioid-related deaths in Colorado, 2000-2015. Am J Public Health 107:1827–1829PubMedCrossRef Livingston MD, Barnett TE, Delcher C, Wagenaar AC (2017) Recreational cannabis legalization and opioid-related deaths in Colorado, 2000-2015. Am J Public Health 107:1827–1829PubMedCrossRef
389.
go back to reference Scavone JL, Sterling RC, Weinstein SP, Van Bockstaele EJ (2013) Impact of cannabis use during stabilization on methadone maintenance treatment. Am J Addict 22:344–351PubMedPubMedCentralCrossRef Scavone JL, Sterling RC, Weinstein SP, Van Bockstaele EJ (2013) Impact of cannabis use during stabilization on methadone maintenance treatment. Am J Addict 22:344–351PubMedPubMedCentralCrossRef
390.
go back to reference Raby WN, Carpenter KM, Rothenberg J et al (2009) Intermittent marijuana use is associated with improved retention in naltrexone treatment for opiate-dependence. Am J Addict 18:301–308PubMedPubMedCentralCrossRef Raby WN, Carpenter KM, Rothenberg J et al (2009) Intermittent marijuana use is associated with improved retention in naltrexone treatment for opiate-dependence. Am J Addict 18:301–308PubMedPubMedCentralCrossRef
391.
go back to reference Bachhuber MA, Saloner B, Cunningham CO, Barry CL (2014) Medical cannabis laws and opioid analgesic overdose mortality in the United States, 1999-2010. JAMA Intern Med 174:1668–1673PubMedPubMedCentralCrossRef Bachhuber MA, Saloner B, Cunningham CO, Barry CL (2014) Medical cannabis laws and opioid analgesic overdose mortality in the United States, 1999-2010. JAMA Intern Med 174:1668–1673PubMedPubMedCentralCrossRef
Metadata
Title
Patterns of medicinal cannabis use, strain analysis, and substitution effect among patients with migraine, headache, arthritis, and chronic pain in a medicinal cannabis cohort
Authors
Eric P. Baron
Philippe Lucas
Joshua Eades
Olivia Hogue
Publication date
01-12-2018
Publisher
Springer Milan
Published in
The Journal of Headache and Pain / Issue 1/2018
Print ISSN: 1129-2369
Electronic ISSN: 1129-2377
DOI
https://doi.org/10.1186/s10194-018-0862-2

Other articles of this Issue 1/2018

The Journal of Headache and Pain 1/2018 Go to the issue