Skip to main content
Top
Published in: Drugs 6/2000

01-12-2000 | Review Article

Cannabinoids in Clinical Practice

Authors: Dr Elizabeth M. Williamson, Fred J. Evans

Published in: Drugs | Issue 6/2000

Login to get access

Abstract

Cannabis has a potential for clinical use often obscured by unreliable and purely anecdotal reports. The most important natural cannabinoid is the psychoactive tetrahydrocannabinol (Δ9-THC); others include cannabidiol (CBD) and cannabigerol (CBG). Not all the observed effects can be ascribed to THC, and the other constituents may also modulate its action; for example CBD reduces anxiety induced by THC. A standardised extract of the herb may be therefore be more beneficial in practice and clinical trial protocols have been drawn up to assess this. The mechanism of action is still not fully understood, although cannabinoid receptors have been cloned and natural ligands identified.
Cannabis is frequently used by patients with multiple sclerosis (MS) for muscle spasm and pain, and in an experimental model of MS low doses of cannabinoids alleviated tremor. Most of the controlled studies have been carried out with THC rather than cannabis herb and so do not mimic the usual clincal situation. Small clinical studies have confirmed the usefulness of THC as an analgesic; CBD and CBG also have analgesic and antiinflammatory effects, indicating that there is scope for developing drugs which do not have the psychoactive properties ofTHC. Patients taking the synthetic derivative nabilone for neurogenic pain actually preferred cannabis herb and reported that it relieved not only pain but the associated depression and anxiety. Cannabinoids are effective in chemotherapy-induced emesis and nabilone has been licensed for this use for several years. Currently, the synthetic cannabinoid HU211 is undergoing trials as a protective agent after brain trauma. Anecdotal reports of cannabis use include case studies in migraine and Tourette’s syndrome, and as a treatment for asthma and glaucoma.
Apart from the smoking aspect, the safety profile of cannabis is fairly good. However, adverse reactions include panic or anxiety attacks, which are worse in the elderly and in women, and less likely in children. Although psychosis has been cited as a consequence of cannabis use, an examination of psychiatric hospital admissions found no evidence of this, however, it may exacerbate existing symptoms. The relatively slow elimination from the body of the cannabinoids has safety implications for cognitive tasks, especially driving and operating machinery; although driving impairment with cannabis is only moderate, there is a significant interaction with alcohol.
Natural materials are highly variable and multiple components need to be standardised to ensure reproducible effects. Pure natural and synthetic compounds do not have these disadvantages but may not have the overall therapeutic effect of the herb.
Literature
1.
go back to reference Formukong EA, Evans AT, Evans FJ. The medicinal uses of cannabis and its constituents. Phytother Res 1989; 3(6): 219–31CrossRef Formukong EA, Evans AT, Evans FJ. The medicinal uses of cannabis and its constituents. Phytother Res 1989; 3(6): 219–31CrossRef
2.
go back to reference Mechoulam R, Ben-Shabat S. From gan-zi-gun-nu to anandamide and 2 — arachidonoylglycerol: the ongoing story of cannabis. Nat Prod Rep 1999; 16: 131–43PubMedCrossRef Mechoulam R, Ben-Shabat S. From gan-zi-gun-nu to anandamide and 2 — arachidonoylglycerol: the ongoing story of cannabis. Nat Prod Rep 1999; 16: 131–43PubMedCrossRef
3.
go back to reference Evans FJ. The medicinal chemistry of cannabis: O’S-haughnessy’s legacy. Pharmaceutical Sci 1997; 3: 533–7 Evans FJ. The medicinal chemistry of cannabis: O’S-haughnessy’s legacy. Pharmaceutical Sci 1997; 3: 533–7
4.
go back to reference Gurley RJ, Aranow R, Katz M. Medicinal marijuana: a review. J Psychoactive Drugs 1998; 30(2): 37–147CrossRef Gurley RJ, Aranow R, Katz M. Medicinal marijuana: a review. J Psychoactive Drugs 1998; 30(2): 37–147CrossRef
5.
go back to reference Hirst RA, Lambert DG, Notcutt WG. Pharmacology and potential therapeutic uses of cannabis. Br J Anaesth 1998; 81: 77–84PubMedCrossRef Hirst RA, Lambert DG, Notcutt WG. Pharmacology and potential therapeutic uses of cannabis. Br J Anaesth 1998; 81: 77–84PubMedCrossRef
6.
go back to reference Pertwee RG. Cannabis and cannabinoids: pharmacology and rationale for clinical use. Pharmaceutical Sci 1997; 3: 539–45 Pertwee RG. Cannabis and cannabinoids: pharmacology and rationale for clinical use. Pharmaceutical Sci 1997; 3: 539–45
7.
go back to reference Fairbairn JW, Hindmarch I, Simic S, et al. Cannabinoid content of some English reefers. Nature 1974; 249: 277–8CrossRef Fairbairn JW, Hindmarch I, Simic S, et al. Cannabinoid content of some English reefers. Nature 1974; 249: 277–8CrossRef
8.
go back to reference De Witt H, Wachtel S. Comparison of whole plant marijuana and Δ9THC in human volunteers. Symposium on the Cannabinoids; 1999 Jun 18–20: Acapulco. Burlington (VT): International Cannabinoid Research Society, 1999: 74 De Witt H, Wachtel S. Comparison of whole plant marijuana and Δ9THC in human volunteers. Symposium on the Cannabinoids; 1999 Jun 18–20: Acapulco. Burlington (VT): International Cannabinoid Research Society, 1999: 74
9.
go back to reference Petitet F, Jeantaud B, Rebaude M, et al. Complex pharmacology of natural cannabinoids: Evidence for partial agonist activity of Δ9 tetrahydrocannabinol and antagonist activity of canna-bidiol on rat brain cannabinoid receptors. Life Sci 1998; 63; PL1–6PubMedCrossRef Petitet F, Jeantaud B, Rebaude M, et al. Complex pharmacology of natural cannabinoids: Evidence for partial agonist activity of Δ9 tetrahydrocannabinol and antagonist activity of canna-bidiol on rat brain cannabinoid receptors. Life Sci 1998; 63; PL1–6PubMedCrossRef
10.
go back to reference Zuardi AW, Shirakawa I, Finkelfarb E, et al. Action of cannabidiol on the anxiety and other effects produced by delta-9-THC in normal subjects. Psychopharmacology 1982; 76: 245–50PubMedCrossRef Zuardi AW, Shirakawa I, Finkelfarb E, et al. Action of cannabidiol on the anxiety and other effects produced by delta-9-THC in normal subjects. Psychopharmacology 1982; 76: 245–50PubMedCrossRef
11.
go back to reference Bornhein LM, Reid M. Influence of cannabinoids on brain levels of other drugs. Symposium on the Cannabinoids; 1999 Jun 18–20: Acapulco. Burlington (VT): International Cannabinoid Research Society, 1999: 84 Bornhein LM, Reid M. Influence of cannabinoids on brain levels of other drugs. Symposium on the Cannabinoids; 1999 Jun 18–20: Acapulco. Burlington (VT): International Cannabinoid Research Society, 1999: 84
12.
go back to reference Working Party of the Pharmaceutical Society of Great Britain. Working Party on the therapeutic uses of cannabis: launch of the protocols for the clinical trials of cannabinoids. London: The Pharmaceutical Society of Great Britain, Jan 1999 Working Party of the Pharmaceutical Society of Great Britain. Working Party on the therapeutic uses of cannabis: launch of the protocols for the clinical trials of cannabinoids. London: The Pharmaceutical Society of Great Britain, Jan 1999
13.
go back to reference Llamas R, Hart R, Schneider N. Allergic bronchopulmonary aspergillosis associated with smoking moldy marijuana. Chest 1978; 6: 871–2CrossRef Llamas R, Hart R, Schneider N. Allergic bronchopulmonary aspergillosis associated with smoking moldy marijuana. Chest 1978; 6: 871–2CrossRef
14.
go back to reference Matsuda LA, Lolait SJ, Brownstein MJ, et al. Structure of a cannabinoid receptor and functional expression of the cloned DNA. Nature 1990; 346: 561–4PubMedCrossRef Matsuda LA, Lolait SJ, Brownstein MJ, et al. Structure of a cannabinoid receptor and functional expression of the cloned DNA. Nature 1990; 346: 561–4PubMedCrossRef
15.
go back to reference Devane WA, Hanus L, Breuer A, et al. Isolation and structure of a brain constituent that binds to the cannabinoid receptor. Science 1992; 258: 1946–9PubMedCrossRef Devane WA, Hanus L, Breuer A, et al. Isolation and structure of a brain constituent that binds to the cannabinoid receptor. Science 1992; 258: 1946–9PubMedCrossRef
16.
go back to reference Monroe S, Thomas KL, Abu-Shaar M. Molecular characterisation of a peripheral receptor for cannabinoids. Nature 1993; 365: 61–5CrossRef Monroe S, Thomas KL, Abu-Shaar M. Molecular characterisation of a peripheral receptor for cannabinoids. Nature 1993; 365: 61–5CrossRef
17.
go back to reference Mechoulam R, Ben-Shabat S, Hanus L, et al. Identification of an endogenous 2-monoglyceride, present in canine gut, that bonds to cannabinoid receptors. Biochem Pharmacol 1995; 50: 83–90PubMedCrossRef Mechoulam R, Ben-Shabat S, Hanus L, et al. Identification of an endogenous 2-monoglyceride, present in canine gut, that bonds to cannabinoid receptors. Biochem Pharmacol 1995; 50: 83–90PubMedCrossRef
18.
go back to reference Mechoulam R, Fride, E, Hanus L, et al. Anandamide may mediate sleep induction. Nature 1997; 389: 25–6PubMedCrossRef Mechoulam R, Fride, E, Hanus L, et al. Anandamide may mediate sleep induction. Nature 1997; 389: 25–6PubMedCrossRef
19.
go back to reference Randall MD, McCulloch AI, Kendall DA. Comparative pharmacology of endothelium-derived hyperpolarizing factor and anandamide in rat isolated mesentery. Eur J Pharmacol 1997; 333(2–3): 191–7PubMedCrossRef Randall MD, McCulloch AI, Kendall DA. Comparative pharmacology of endothelium-derived hyperpolarizing factor and anandamide in rat isolated mesentery. Eur J Pharmacol 1997; 333(2–3): 191–7PubMedCrossRef
20.
go back to reference Fride E, Barg J, Levy D, et al. Low doses of anandamide inhibit pharmacological effects of Δ9 tetrahydrocannabinol. J Pharmacol Exp Ther 1995; 272: 699–707PubMed Fride E, Barg J, Levy D, et al. Low doses of anandamide inhibit pharmacological effects of Δ9 tetrahydrocannabinol. J Pharmacol Exp Ther 1995; 272: 699–707PubMed
21.
go back to reference Di Marzo V, Melck D, Bosogno T, et al. Endocannabinoids: endogenous cannabinoid receptor ligands with neuromodulatory action. Trends Neurosci 1998; 21(12): 521–8PubMedCrossRef Di Marzo V, Melck D, Bosogno T, et al. Endocannabinoids: endogenous cannabinoid receptor ligands with neuromodulatory action. Trends Neurosci 1998; 21(12): 521–8PubMedCrossRef
22.
go back to reference Consroe P. Brain cannabinoid systems as targets for the therapy of neurological disorders. Neurobiol Dis 1998; 5: 534–51PubMedCrossRef Consroe P. Brain cannabinoid systems as targets for the therapy of neurological disorders. Neurobiol Dis 1998; 5: 534–51PubMedCrossRef
23.
go back to reference Evans FJ. Cannabinoids: the separation of central from peripheral effects on a structural basis. Planta Med 1991; 57 Suppl.: 60–7CrossRef Evans FJ. Cannabinoids: the separation of central from peripheral effects on a structural basis. Planta Med 1991; 57 Suppl.: 60–7CrossRef
24.
go back to reference Formukong E, Garland LG, Evans AT, et al. Inhibition of A23187 induced release of CTB4 in mouse blood in vivo and human polymorphonuclear cells in vitro by analgesic cannabidiol. Phytother Res 1991; 5: 258–61CrossRef Formukong E, Garland LG, Evans AT, et al. Inhibition of A23187 induced release of CTB4 in mouse blood in vivo and human polymorphonuclear cells in vitro by analgesic cannabidiol. Phytother Res 1991; 5: 258–61CrossRef
25.
go back to reference Morgan DR, editor. Therapeutic uses of cannabis: British Medical Association. Netherlands: Harwood Academic Publishers, 1997 Morgan DR, editor. Therapeutic uses of cannabis: British Medical Association. Netherlands: Harwood Academic Publishers, 1997
26.
go back to reference Select Committee on Science and Technology. Cannabis: the scientific and medical evidence. The House of Lords Session 1997–8. 9th report [HL paper 151]. London: The Stationary Office, 1998 Select Committee on Science and Technology. Cannabis: the scientific and medical evidence. The House of Lords Session 1997–8. 9th report [HL paper 151]. London: The Stationary Office, 1998
27.
28.
go back to reference Brenneisen R, Egli MA, Elsohly V, et al. The effect of orally and rectally administered delta-9-THC on spasticity: a pilot study with two patients. Int J Clin Pharmacol Ther 1996; 34: 446–52PubMed Brenneisen R, Egli MA, Elsohly V, et al. The effect of orally and rectally administered delta-9-THC on spasticity: a pilot study with two patients. Int J Clin Pharmacol Ther 1996; 34: 446–52PubMed
29.
go back to reference Ungerleider JT, Andyrsiak T, Fairbanks L, et al. Δ9-THC in the treatment of spasticity associated with multiple sclerosis. Adv Alcohol Subst Abuse 1987; 7: 39–50PubMedCrossRef Ungerleider JT, Andyrsiak T, Fairbanks L, et al. Δ9-THC in the treatment of spasticity associated with multiple sclerosis. Adv Alcohol Subst Abuse 1987; 7: 39–50PubMedCrossRef
30.
go back to reference Consroe P, Musty R, Rein R, et al. The perceived effects of smoked cannabis on patients with MS. Eur Neurol 1997; 38: 44–7PubMedCrossRef Consroe P, Musty R, Rein R, et al. The perceived effects of smoked cannabis on patients with MS. Eur Neurol 1997; 38: 44–7PubMedCrossRef
31.
go back to reference Schon F, Hart P, Hodgson TR, et al. Suppression of Pendular Nystagmus by cannabis in a patient with multiple sclerosis. Neurology 1999; 53: 2209–10PubMedCrossRef Schon F, Hart P, Hodgson TR, et al. Suppression of Pendular Nystagmus by cannabis in a patient with multiple sclerosis. Neurology 1999; 53: 2209–10PubMedCrossRef
32.
go back to reference Willis S. The use of cannabis in multiple sclerosis. Pharm J 1995; 255: 237–8 Willis S. The use of cannabis in multiple sclerosis. Pharm J 1995; 255: 237–8
33.
go back to reference Formukong EA, Evans AT, Evans FJ. Inhibition of the cataleptic effects of Δ9-THC by other constituents of Cannabis sativa. J Pharm Pharmacol 1988; 40: 132–43PubMedCrossRef Formukong EA, Evans AT, Evans FJ. Inhibition of the cataleptic effects of Δ9-THC by other constituents of Cannabis sativa. J Pharm Pharmacol 1988; 40: 132–43PubMedCrossRef
34.
go back to reference Martyn CN, Illis LS, Thorn J. Nabilone in the treatment of multiple sclerosis. Lancet 1995; 345: 579PubMedCrossRef Martyn CN, Illis LS, Thorn J. Nabilone in the treatment of multiple sclerosis. Lancet 1995; 345: 579PubMedCrossRef
35.
go back to reference Greenberg HS, Werness SAS, Pugh JE, et al. Short-term effects of smoking marihuana on balance in patients with multiple sclerosis and normal volunteers. Clin Pharmacol Ther 1994; 55: 324–8PubMedCrossRef Greenberg HS, Werness SAS, Pugh JE, et al. Short-term effects of smoking marihuana on balance in patients with multiple sclerosis and normal volunteers. Clin Pharmacol Ther 1994; 55: 324–8PubMedCrossRef
36.
go back to reference Meinck H, Schonle P, Conrad B. Effect of cannabinoids on spasticity and ataxia in multiple sclerosis. J Neurol 1989; 236: 120–2PubMedCrossRef Meinck H, Schonle P, Conrad B. Effect of cannabinoids on spasticity and ataxia in multiple sclerosis. J Neurol 1989; 236: 120–2PubMedCrossRef
37.
go back to reference Consroe P, Sandyk R, Snider SR. Open label evaluation of cannabidiol in dystonic movement disorders. Int J Neurosci 1986; 30: 277–82PubMedCrossRef Consroe P, Sandyk R, Snider SR. Open label evaluation of cannabidiol in dystonic movement disorders. Int J Neurosci 1986; 30: 277–82PubMedCrossRef
38.
go back to reference Sandyk R, Snider SR, Consroe P, et al. Cannabidiol in dystonic movement disorders [letter]. Psychiatry Res 1986; 18(3): 291PubMedCrossRef Sandyk R, Snider SR, Consroe P, et al. Cannabidiol in dystonic movement disorders [letter]. Psychiatry Res 1986; 18(3): 291PubMedCrossRef
39.
go back to reference Snider SR, Consroe P. Treatment of Meige’s syndrome with cannabidiol. Neurology 1984; 34 Suppl.: 147 Snider SR, Consroe P. Treatment of Meige’s syndrome with cannabidiol. Neurology 1984; 34 Suppl.: 147
40.
go back to reference Muller-Vahl K, Schneider U, Emrich HM. Nabilone increases choreatic movements in Huntingdon’s disease. Mov Disord 1999; 14(6): 1038–40PubMedCrossRef Muller-Vahl K, Schneider U, Emrich HM. Nabilone increases choreatic movements in Huntingdon’s disease. Mov Disord 1999; 14(6): 1038–40PubMedCrossRef
41.
go back to reference Frankel JP, Hughes A, Lees AJ, et al. Marijuana for Parkinsonian tremor [letter]. J Neurol Neurosurg Psychiatry 1990; 53: 436PubMedCrossRef Frankel JP, Hughes A, Lees AJ, et al. Marijuana for Parkinsonian tremor [letter]. J Neurol Neurosurg Psychiatry 1990; 53: 436PubMedCrossRef
42.
go back to reference Noyes R, Brunk ST, Avery DH, et al. The analgesic properties of Δ9-THC and codeine. Clin Pharmacol Ther 1975; 18: 84–9PubMed Noyes R, Brunk ST, Avery DH, et al. The analgesic properties of Δ9-THC and codeine. Clin Pharmacol Ther 1975; 18: 84–9PubMed
43.
go back to reference Holdcroft A, Smith M, Jacklin A, et al. Pain relief with oral cannabinoids in familial Mediterranean fever. Anaesthesia 1997; 52: 483–8PubMedCrossRef Holdcroft A, Smith M, Jacklin A, et al. Pain relief with oral cannabinoids in familial Mediterranean fever. Anaesthesia 1997; 52: 483–8PubMedCrossRef
44.
go back to reference Notcutt WG, Price M, Chapman G. Clinical experience with nabilone for chronic pain. Pharm Sci 1997; 3: 551–5 Notcutt WG, Price M, Chapman G. Clinical experience with nabilone for chronic pain. Pharm Sci 1997; 3: 551–5
45.
go back to reference Formukong EA, Evans AT, Evans FJ. Analgesic and anti-inflammatory activity of the constituents of Cannabis sativa. Inflammation 1988; 12: 361–71PubMedCrossRef Formukong EA, Evans AT, Evans FJ. Analgesic and anti-inflammatory activity of the constituents of Cannabis sativa. Inflammation 1988; 12: 361–71PubMedCrossRef
47.
go back to reference Formukong EA, Evans AT, Evans FJ. The inhibitory effects of cannabinoids, the active constituents of Cannabis sativa on human and rabbit platelet aggregation. J Pharm Pharmacol 1989; 41: 705–9PubMedCrossRef Formukong EA, Evans AT, Evans FJ. The inhibitory effects of cannabinoids, the active constituents of Cannabis sativa on human and rabbit platelet aggregation. J Pharm Pharmacol 1989; 41: 705–9PubMedCrossRef
48.
go back to reference Martin BR, Lichtman AH. Cannabinoid transmission and pain perception. Neurobiol Dis 1998; 5: 447–61PubMedCrossRef Martin BR, Lichtman AH. Cannabinoid transmission and pain perception. Neurobiol Dis 1998; 5: 447–61PubMedCrossRef
49.
go back to reference Hill SY, Schwin R, Goodwin DW, et al. Marijuana and pain. J Pharmacol Exp Ther 1974; 188: 415–18PubMed Hill SY, Schwin R, Goodwin DW, et al. Marijuana and pain. J Pharmacol Exp Ther 1974; 188: 415–18PubMed
50.
go back to reference Clark WC, Janal MN, Zeidenberg P, et al. Effects of moderate and high doses of marihuana on thermal pain: a sensory decision theory analysis. J Clin Pharmacol 1981; 21: 299S–310PubMed Clark WC, Janal MN, Zeidenberg P, et al. Effects of moderate and high doses of marihuana on thermal pain: a sensory decision theory analysis. J Clin Pharmacol 1981; 21: 299S–310PubMed
51.
go back to reference Hampson AJ, Grimaldi M, Axelrod J, et al. Cannabidiol and ((−)-Δ9 tetrahydrocannabinol are neuroprotective antioxidants. Proc Natl Acad Sci U S A 1998; 95: 8268–73PubMedCrossRef Hampson AJ, Grimaldi M, Axelrod J, et al. Cannabidiol and ((−)-Δ9 tetrahydrocannabinol are neuroprotective antioxidants. Proc Natl Acad Sci U S A 1998; 95: 8268–73PubMedCrossRef
52.
go back to reference French ED, Dillon K, Wu X. Cannabinoids excite dopamine neurons in the ventral tegmentum and substantia nigra. Neuroreport 1997; 8: 649–52PubMedCrossRef French ED, Dillon K, Wu X. Cannabinoids excite dopamine neurons in the ventral tegmentum and substantia nigra. Neuroreport 1997; 8: 649–52PubMedCrossRef
53.
54.
go back to reference Hemming M, Yellowlees PM. Effective treatment of Tourett’s syndrome with marijuana. J Psychopharmacol 1993; 7: 389–91PubMedCrossRef Hemming M, Yellowlees PM. Effective treatment of Tourett’s syndrome with marijuana. J Psychopharmacol 1993; 7: 389–91PubMedCrossRef
55.
go back to reference Muller-Vahl KR, Kolbe H, Schneider U, et al. Cannabinoids: possible role in pathophysiology and therapy of Gilles de la Tourette syndrome. Acta Psychiatrica Scand 1998; 98(6): 502–6CrossRef Muller-Vahl KR, Kolbe H, Schneider U, et al. Cannabinoids: possible role in pathophysiology and therapy of Gilles de la Tourette syndrome. Acta Psychiatrica Scand 1998; 98(6): 502–6CrossRef
56.
go back to reference Muller-Vahl KR, Kolbe H, Dengler R. Gilles de la Tourette syndrome: influence of nicotine, alcohol and marijuana on the classical symptoms. Nervenartz 1997; 68(12): 985–9CrossRef Muller-Vahl KR, Kolbe H, Dengler R. Gilles de la Tourette syndrome: influence of nicotine, alcohol and marijuana on the classical symptoms. Nervenartz 1997; 68(12): 985–9CrossRef
57.
go back to reference Muller-Vahl K, Schneider U, Kolbe H, et al. Treatment of Tourette’s syndrome with delta-9-tetrahydrocannabinol. Am J Psychiatry 1999; 156(3): 495PubMed Muller-Vahl K, Schneider U, Kolbe H, et al. Treatment of Tourette’s syndrome with delta-9-tetrahydrocannabinol. Am J Psychiatry 1999; 156(3): 495PubMed
58.
go back to reference Gruber AJ, Pope HG, Brown ME. Do patients use marijuana as an antidepressant? Depression 1996; 4(2): 77–80PubMedCrossRef Gruber AJ, Pope HG, Brown ME. Do patients use marijuana as an antidepressant? Depression 1996; 4(2): 77–80PubMedCrossRef
59.
go back to reference Fishman SM, Rosenbaum JF, Yabusaki DI, et al. Marijuana-related anxiety: a questionnaire based pilot study of normal and psychiatric populations. Res Comm Subst Abuse 1988; 9(3–4): 219–26 Fishman SM, Rosenbaum JF, Yabusaki DI, et al. Marijuana-related anxiety: a questionnaire based pilot study of normal and psychiatric populations. Res Comm Subst Abuse 1988; 9(3–4): 219–26
60.
go back to reference Musty RE, Kaback L. Relationship between motivation and depression in chronic marijuana users. Life Sci 1995; 56(23–24): 2151–8PubMedCrossRef Musty RE, Kaback L. Relationship between motivation and depression in chronic marijuana users. Life Sci 1995; 56(23–24): 2151–8PubMedCrossRef
61.
go back to reference Grinspoon L, Bahalar JB. The use of cannabis as a mood stabilizer in bipolar disorder: anecdotal evidence and the need for clinical research. J Psychoactive Drugs 1998; 30(2): 171–7PubMedCrossRef Grinspoon L, Bahalar JB. The use of cannabis as a mood stabilizer in bipolar disorder: anecdotal evidence and the need for clinical research. J Psychoactive Drugs 1998; 30(2): 171–7PubMedCrossRef
62.
go back to reference Gruber A, Pope H. Cannabis psychotic disorder: does it exist? Am J Addict 1994; 3: 72–83 Gruber A, Pope H. Cannabis psychotic disorder: does it exist? Am J Addict 1994; 3: 72–83
63.
go back to reference Pope H, Gruber A, Yurgelan-Todd D. The residual neuropsychological effects of cannabis: the current status of research. Drug Alcohol Depend 1995; 38: 25–34PubMedCrossRef Pope H, Gruber A, Yurgelan-Todd D. The residual neuropsychological effects of cannabis: the current status of research. Drug Alcohol Depend 1995; 38: 25–34PubMedCrossRef
64.
go back to reference Ries R K. The dually diagnosed patient with psychotic symptoms. J Addict Dis 1993; 12(3): 103–122PubMedCrossRef Ries R K. The dually diagnosed patient with psychotic symptoms. J Addict Dis 1993; 12(3): 103–122PubMedCrossRef
65.
go back to reference Leweke FM, Guiffrida A, Wurster U, et al. Elevated endogenous cannabinoids in schizophrenia. Neuroreport. 1999; 10(8): 1665–9PubMedCrossRef Leweke FM, Guiffrida A, Wurster U, et al. Elevated endogenous cannabinoids in schizophrenia. Neuroreport. 1999; 10(8): 1665–9PubMedCrossRef
67.
go back to reference Zuardi AW, Rodrigues J, Cunha JM. Effects of cannabidiol in animal models predictive of antipsychotic activity. Psycho-pharmacology 1991; 104(2): 260–4 Zuardi AW, Rodrigues J, Cunha JM. Effects of cannabidiol in animal models predictive of antipsychotic activity. Psycho-pharmacology 1991; 104(2): 260–4
68.
go back to reference Zuardi AW, Morais SL, Guimares FS, et al. Antipsychotic effect of cannabidiol. J Clin Psychiatry 1995; 56(10): 485–6PubMed Zuardi AW, Morais SL, Guimares FS, et al. Antipsychotic effect of cannabidiol. J Clin Psychiatry 1995; 56(10): 485–6PubMed
69.
go back to reference Wagner J, Varga K, Kunos G. Cardiovascular actions of the cannabinoids and their generation during shock. J Mol Med 1998; 76: 824–36PubMedCrossRef Wagner J, Varga K, Kunos G. Cardiovascular actions of the cannabinoids and their generation during shock. J Mol Med 1998; 76: 824–36PubMedCrossRef
70.
go back to reference Adams MD, Earnhard JT, Martin BR, et al. Acannabinoid with cardiovascular activity but no overt behavioural effects. Experientia 1977; 33: 1204–5PubMedCrossRef Adams MD, Earnhard JT, Martin BR, et al. Acannabinoid with cardiovascular activity but no overt behavioural effects. Experientia 1977; 33: 1204–5PubMedCrossRef
71.
72.
go back to reference Green K, Wynn H, Bowman KA. A comparison of topical cannabinoids on the intraocular pressure. Exp Eye Res 1978; 27: 239–46PubMedCrossRef Green K, Wynn H, Bowman KA. A comparison of topical cannabinoids on the intraocular pressure. Exp Eye Res 1978; 27: 239–46PubMedCrossRef
73.
go back to reference Maor D, Trevess T, Korczyn AD. Lack of effect of cannabinoids on carbonic anhydrase. J Neural Transm 1980; 49: 205–6PubMedCrossRef Maor D, Trevess T, Korczyn AD. Lack of effect of cannabinoids on carbonic anhydrase. J Neural Transm 1980; 49: 205–6PubMedCrossRef
74.
go back to reference Merritt JC, Perry DD, Russell DN, et al. Topical Δ9 tetrahydrocannabinol and aqueous humor dynamics in glaucoma. J Clin Pharmacol 1981; 21 (8–9 Suppl.): 467S–71PubMed Merritt JC, Perry DD, Russell DN, et al. Topical Δ9 tetrahydrocannabinol and aqueous humor dynamics in glaucoma. J Clin Pharmacol 1981; 21 (8–9 Suppl.): 467S–71PubMed
75.
go back to reference Green K. Marijuana smoking vs cannabinoids for glaucoma therapy. Arch Ophthalmol 1998; 116(11): 433–1437 Green K. Marijuana smoking vs cannabinoids for glaucoma therapy. Arch Ophthalmol 1998; 116(11): 433–1437
76.
go back to reference Chang A, Shiling D, Stillman R, et al. Δ9 tetrahydrocannabinol as an antiemetic in cancer patients receiving high dose methotrexate. Ann Intern Med 1979; 91: 819–24PubMed Chang A, Shiling D, Stillman R, et al. Δ9 tetrahydrocannabinol as an antiemetic in cancer patients receiving high dose methotrexate. Ann Intern Med 1979; 91: 819–24PubMed
77.
go back to reference McCabe M, Smith F, Macdonald J, et al. Efficacy of tetrahydrocannabinol in patients refractory to standard antiemetic therapy. Invest New Drugs 1988; 6: 243–6PubMedCrossRef McCabe M, Smith F, Macdonald J, et al. Efficacy of tetrahydrocannabinol in patients refractory to standard antiemetic therapy. Invest New Drugs 1988; 6: 243–6PubMedCrossRef
78.
go back to reference Tortorice P, O’Connell M. Management of chemotherapy-induced nausea and vomiting. Pharmacotherapy 1990; 10(2): 129–45PubMed Tortorice P, O’Connell M. Management of chemotherapy-induced nausea and vomiting. Pharmacotherapy 1990; 10(2): 129–45PubMed
79.
go back to reference Cunningham D, Bradley C, Forrest G, et al. 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 analogs. Eur J Cancer Clin Oncol 1988; 24: 685–9PubMedCrossRef Cunningham D, Bradley C, Forrest G, et al. 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 analogs. Eur J Cancer Clin Oncol 1988; 24: 685–9PubMedCrossRef
80.
go back to reference Abrahamov A, Abrahamov A, Mechoulam R. An efficient new cannabinoid antiemetic in pediatric oncology. Life Sci 1995; 56(23–24): 2097–102PubMedCrossRef Abrahamov A, Abrahamov A, Mechoulam R. An efficient new cannabinoid antiemetic in pediatric oncology. Life Sci 1995; 56(23–24): 2097–102PubMedCrossRef
81.
go back to reference Cat L, Coleman D. DIAS rounds: treatment for HIV wasting syndrome. Ann Pharmacother 1994; 28: 505–7 Cat L, Coleman D. DIAS rounds: treatment for HIV wasting syndrome. Ann Pharmacother 1994; 28: 505–7
82.
go back to reference Plasse TF, Gorter RW, Krasnow SH, et al. Recent clinical experience with dronabinol. Pharmacol Biochem Behav 1991; 40: 695–700PubMedCrossRef Plasse TF, Gorter RW, Krasnow SH, et al. Recent clinical experience with dronabinol. Pharmacol Biochem Behav 1991; 40: 695–700PubMedCrossRef
83.
go back to reference Beale JE, Olson R, Laubenstein L, et al. J Pain Symptom Manage 1995; 10(2): 89–97CrossRef Beale JE, Olson R, Laubenstein L, et al. J Pain Symptom Manage 1995; 10(2): 89–97CrossRef
84.
go back to reference Abboud R, Sanders H. Effect of oral administration of delta-9-tetrahydrocannabinol on airway mechanics in normal and asthmatic subjects. Chest 1976; 70: 480–5PubMedCrossRef Abboud R, Sanders H. Effect of oral administration of delta-9-tetrahydrocannabinol on airway mechanics in normal and asthmatic subjects. Chest 1976; 70: 480–5PubMedCrossRef
85.
go back to reference Williams S, Hartley J, Graham J. Bronchodilator effect of delta-9-tetrahydrocannabinol administered by aerosol to asthmatic patients. Thorax 1976; 31: 720–3PubMedCrossRef Williams S, Hartley J, Graham J. Bronchodilator effect of delta-9-tetrahydrocannabinol administered by aerosol to asthmatic patients. Thorax 1976; 31: 720–3PubMedCrossRef
86.
go back to reference Kirk JM, Doty P, De Wit H. Effects of expectancies on subjective responses to oral Δ9 tetrahydrocannabinol. Pharmacol Biochem Behav 1998; 59(2): 287–93PubMedCrossRef Kirk JM, Doty P, De Wit H. Effects of expectancies on subjective responses to oral Δ9 tetrahydrocannabinol. Pharmacol Biochem Behav 1998; 59(2): 287–93PubMedCrossRef
87.
go back to reference Huestis MA, Cone EJ. Urinary excretion half-life of 11-nor-9-carboxy-Δ9-tetrahydrocannabinol in humans. Ther Drug Monit 1998; 20: 570–6PubMedCrossRef Huestis MA, Cone EJ. Urinary excretion half-life of 11-nor-9-carboxy-Δ9-tetrahydrocannabinol in humans. Ther Drug Monit 1998; 20: 570–6PubMedCrossRef
88.
go back to reference Johansson E, Noren K, Sjovall J, et al. Determination of delta-1-tetrahydrocannabinol in human fat biopsies from marihuana users by gas chromatography-mass spectrometry. Biomed Chromatogr 1989; 3: 35–8PubMedCrossRef Johansson E, Noren K, Sjovall J, et al. Determination of delta-1-tetrahydrocannabinol in human fat biopsies from marihuana users by gas chromatography-mass spectrometry. Biomed Chromatogr 1989; 3: 35–8PubMedCrossRef
89.
go back to reference Hunt CA, Jones RT. Tolerance and disposition of tetrahydrocannabinol in man. J Pharmacol Exp Ther 1980; 213: 35–44 Hunt CA, Jones RT. Tolerance and disposition of tetrahydrocannabinol in man. J Pharmacol Exp Ther 1980; 213: 35–44
90.
go back to reference Robbe H. Marijuana’s impairing effects on driving are moderate when taken alone but severe when combined with alcohol. Hum Psychopharmacol Clin Ther 1998; 13: S70–8CrossRef Robbe H. Marijuana’s impairing effects on driving are moderate when taken alone but severe when combined with alcohol. Hum Psychopharmacol Clin Ther 1998; 13: S70–8CrossRef
91.
go back to reference Fant R, Heishman SJ, Bunker EB, et al. Acute and residual effects of marijuana in humans. Pharmacol Biochem Behav 1998; 60(4): 777–84PubMedCrossRef Fant R, Heishman SJ, Bunker EB, et al. Acute and residual effects of marijuana in humans. Pharmacol Biochem Behav 1998; 60(4): 777–84PubMedCrossRef
92.
go back to reference Anon. Cannabis complication. Pharm J 1999; 263: 146–92 Anon. Cannabis complication. Pharm J 1999; 263: 146–92
93.
go back to reference Manzanares J, Corchero J, Romero J, et al. Pharmacological and biochemical interactions between opioids and cannabinoids. Trends Pharm Sci 1999: 20: 287–94PubMedCrossRef Manzanares J, Corchero J, Romero J, et al. Pharmacological and biochemical interactions between opioids and cannabinoids. Trends Pharm Sci 1999: 20: 287–94PubMedCrossRef
Metadata
Title
Cannabinoids in Clinical Practice
Authors
Dr Elizabeth M. Williamson
Fred J. Evans
Publication date
01-12-2000
Publisher
Springer International Publishing
Published in
Drugs / Issue 6/2000
Print ISSN: 0012-6667
Electronic ISSN: 1179-1950
DOI
https://doi.org/10.2165/00003495-200060060-00005

Other articles of this Issue 6/2000

Drugs 6/2000 Go to the issue

Adis New Drug Profile

Lopinavir

Adis New Drug Profile

Lopinavir