[Skip to Content]
Sign In
Individual Sign In
Create an Account
Institutional Sign In
OpenAthens Shibboleth
Purchase Options:
[Skip to Content Landing]
Figure 1.
Flow of Studies Through the Review Process
Flow of Studies Through the Review Process

AE indicates adverse event; RCT, randomized controlled trial; and SR, systematic review.

aThese excluded reports were screened as full-text articles/reports.

bThe number of included RCTS does not sum because some were included in more than 1 indication category.

Figure 2.
Improvement in Pain
Improvement in Pain

Odds indicate 30% or greater improvement in pain with cannabinoid compared with placebo, stratified according to cannabinoid. The square data markers indicate odds ratios (ORs) from primary studies, with sizes reflecting the statistical weight of the study using random-effects meta-analysis. The horizontal lines indicate 95% CIs. The blue diamond data markers represent the subtotal and overall OR and 95% CI. The vertical dashed line shows the summary effect estimate, the dotted shows the line of no effect (OR = 1).

Figure 3.
Change in Ashworth Score for Cannabinoid Compared With Placebo, Stratified According to Cannabinoid
Change in Ashworth Score for Cannabinoid Compared With Placebo, Stratified According to Cannabinoid

The square data markers indicate mean differences from primary studies, with sizes reflecting the statistical weight of the study using random-effects meta-analysis. The horizontal line indicate, 95% CIs. The blue diamond data markers represent the subtotal and overall weighted mean difference and 95% CI. The vertical dashed line shows the summary effect estimate, the solid vertical line shows the line of no effect (mean difference = 0).

Figure 4.
Odds of Having Any Adverse Event With Cannabinoids Compared With Placebo, Stratified According to Cannabinoid
Odds of Having Any Adverse Event With Cannabinoids Compared With Placebo, Stratified According to Cannabinoid

The square data markers indicate odds ratios (ORs) from primary studies, with sizes reflecting the statistical weight of the study using random-effects meta-analysis. The horizontal lines indicate 95% CIs. The blue diamond data markers represent the subtotal and overall OR and 95% CI. The vertical dashed line shows the summary effect estimate, the dotted line shows the line of no effect (OR = 1).

Table 1.  
Evaluation of Interventions by Included Studies
Evaluation of Interventions by Included Studies
Table 2.  
Summary Estimates From Meta-analyses of Parallel-Group Studies and Results for Primary Outcomes With Associated GRADE Ratings
Summary Estimates From Meta-analyses of Parallel-Group Studies and Results for Primary Outcomes With Associated GRADE Ratings
Table 3.  
Summary Estimates From Meta-analyses for Each AE Assessed: Odds of Participants Experiencing AE With Cannabinoid vs Placebo or Active Comparison
Summary Estimates From Meta-analyses for Each AE Assessed: Odds of Participants Experiencing AE With Cannabinoid vs Placebo or Active Comparison
1.
Small  E, Cronquist  A.  A practical and natural taxonomy for cannabis.  Taxon. 1976;25(4):405-435. doi:10.2307/1220524.Google ScholarCrossref
2.
Poznyak  V.  SY14-1 global epidemiology of cannabis use and implications for public health.  Alcohol Alcohol. 2014;49(suppl 1):i14. doi:10.1093/alcalc/agu052.58 i.Google ScholarCrossref
3.
United Nations.  Single Convention on Narcotic Drugs, 1961. New York, NY: United Nations; 1962.
4.
Hazekamp  A, Ware  MA, Müller-Vahl  KR, Abrams  D, Grotenhermen  F.  The medicinal use of cannabis and cannabinoids—an international cross-sectional survey on administration forms.  J Psychoactive Drugs. 2013;45(3):199-210.PubMedGoogle ScholarCrossref
5.
Hazekamp  A, Heerdink  ER.  The prevalence and incidence of medicinal cannabis on prescription in the Netherlands.  Eur J Clin Pharmacol. 2013;69(8):1575-1580.PubMedGoogle ScholarCrossref
6.
Office of National Drug Control Policy.  Marijuana Resource Center: State Laws Related to Marijuana.https://www.whitehouse.gov/ondcp/state-laws-related-to-marijuana. Accessed May 18, 2015.
7.
Higgins  JPT, Green  S, eds.  Cochrane Handbook for Systematic Reviews of Interventions. Version 5.1.0 (updated March 2011). The Cochrane Collaboration website. http://handbook.cochrane.org/. Accessed March 23, 2011.
8.
Centre for Reviews and Dissemination.  Systematic Reviews: CRD’s Guidance for Undertaking Reviews in Health Care (Internet). York, England: University of York; 2009. https://www.york.ac.uk/media/crd/Systematic_Reviews.pdf. Accessed March 23, 2011.
9.
Canadian Agency for Drugs and Technologies in Health.  CADTH Peer Review Checklist for Search Strategies (Internet). Ottawa, Canada: CADTH; 2013. https://www.cadth.ca/resources/finding-evidence/cadth-peer-review-checklist-search-strategies. Accessed March 17, 2014.
10.
Bohannon  RW, Smith  MB.  Interrater reliability of a modified Ashworth scale of muscle spasticity.  Phys Ther. 1987;67(2):206-207.PubMedGoogle Scholar
11.
Higgins  JP, Altman  DG, Gøtzsche  PC,  et al; Cochrane Bias Methods Group; Cochrane Statistical Methods Group.  The Cochrane Collaboration’s tool for assessing risk of bias in randomised trials.  BMJ. 2011;343:d5928.PubMedGoogle ScholarCrossref
12.
DerSimonian  R, Laird  N.  Meta-analysis in clinical trials.  Control Clin Trials. 1986;7(3):177-188.PubMedGoogle ScholarCrossref
13.
Higgins  JP, Thompson  SG.  Quantifying heterogeneity in a meta-analysis.  Stat Med. 2002;21(11):1539-1558.PubMedGoogle ScholarCrossref
14.
Guyatt  GH, Oxman  AD, Vist  G,  et al.  GRADE guidelines: 4. rating the quality of evidence—study limitations (risk of bias).  J Clin Epidemiol. 2011;64(4):407-415.PubMedGoogle ScholarCrossref
15.
Broder  LE, Lean  NL, Hilsenbeck  SG.  A randomized blinded clinical trial comparing delta-9-tetrahydrocannabinol (THC) and hydroxizine (HZ) as antiemetics (AE) for cancer chemotherapy (CT).  Proc Am Assoc Cancer Res. 1982;23:514.Google Scholar
16.
Long  A, Mioduszewski  J, Natale  R.  A randomized double-blind cross-over comparison of the antiemetic activity of levonantradol and prochlorperazine.  Proc Am Soc Clin Oncol. 1982;1:C-220.Google Scholar
17.
Hagenbach  U, Luz  S, Brenneisen  R, Mäder  M.  The treatment of spasticity with D9-tetrahydrocannabinol (D9-THC) in patients with spinal cord injury. Paper presented at: IACM 2nd Conference on Cannabinoids in Medicine; September 12-13, 2003; Cologne, Germany.
18.
Prasad  B, Radulovacki  MG, Carley  DW.  Randomized placebo controlled trial of dronabinol in obstructive sleep apnea. Paper presented at: American Thoracic Society International Conference, ATS 2011; May 13-18, 2011; Denver, CO. Am J Respir Crit Care Med. 2011;183(1):A2720.
19.
GW Pharmaceuticals Ltd.  A study of cannabis based medicine extracts and placebo in patients with pain due to spinal cord injury. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT01606202. Accessed April 7, 2014.
20.
Center for Medicinal Cannabis Research.  Efficacy of inhaled cannabis in diabetic painful peripheral neuropathy. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00781001. Accessed April 7, 2014.
21.
Stanley Medical Research Institute, Coordinating Centre for Clinical Trials Cologne.  University of Cologne. A clinical trial on the antipsychotic properties of cannabidiol. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00309413. Accessed April 7, 2014.
22.
GW Pharma Ltd.  A double blind, randomised, placebo controlled, parallel group study of Sativex in the treatment of subjects with pain due to diabetic neuropathy. EU Clinical Trials Register. https://www.clinicaltrialsregister.eu/ctr-search/search?query=eudract_number:2004-002530-20. Accessed August 4, 2014.
23.
GW Pharmaceuticals Ltd.  A study to evaluate the effects of cannabis based medicine in patients with pain of neurological origin. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT01606176 Accessed April 7, 2014.
24.
Duran  M, Pérez  E, Abanades  S,  et al.  Preliminary efficacy and safety of an oromucosal standardized cannabis extract in chemotherapy-induced nausea and vomiting.  Br J Clin Pharmacol. 2010;70(5):656-663.PubMedGoogle ScholarCrossref
25.
Meiri  E, Jhangiani  H, Vredenburgh  JJ,  et al.  Efficacy of dronabinol alone and in combination with ondansetron versus ondansetron alone for delayed chemotherapy-induced nausea and vomiting.  Curr Med Res Opin. 2007;23(3):533-543.PubMedGoogle ScholarCrossref
26.
Lane  M, Vogel  CL, Ferguson  J,  et al.  Dronabinol and prochlorperazine in combination for treatment of cancer chemotherapy-induced nausea and vomiting.  J Pain Symptom Manage. 1991;6(6):352-359.PubMedGoogle ScholarCrossref
27.
McCabe  M, Smith  FP, Macdonald  JS, Woolley  PV, Goldberg  D, Schein  PS.  Efficacy of tetrahydrocannabinol in patients refractory to standard antiemetic therapy.  Invest New Drugs. 1988;6(3):243-246.PubMedGoogle ScholarCrossref
28.
Chan  HS, Correia  JA, MacLeod  SM.  Nabilone versus prochlorperazine for control of cancer chemotherapy-induced emesis in children: a double-blind, crossover trial.  Pediatrics. 1987;79(6):946-952.PubMedGoogle Scholar
29.
Pomeroy  M, Fennelly  JJ, Towers  M.  Prospective randomized double-blind trial of nabilone versus domperidone in the treatment of cytotoxic-induced emesis.  Cancer Chemother Pharmacol. 1986;17(3):285-288.PubMedGoogle ScholarCrossref
30.
Dalzell  AM, Bartlett  H, Lilleyman  JS.  Nabilone: an alternative antiemetic for cancer chemotherapy.  Arch Dis Child. 1986;61(5):502-505.PubMedGoogle ScholarCrossref
31.
Niederle  N, Schütte  J, Schmidt  CG.  Crossover comparison of the antiemetic efficacy of nabilone and alizapride in patients with nonseminomatous testicular cancer receiving cisplatin therapy.  Klin Wochenschr. 1986;64(8):362-365.PubMedGoogle ScholarCrossref
32.
Niiranen  A, Mattson  K.  A cross-over comparison of nabilone and prochlorperazine for emesis induced by cancer chemotherapy.  Am J Clin Oncol. 1985;8(4):336-340.PubMedGoogle ScholarCrossref
33.
Heim  ME, Queisser  W, Altenburg  HP.  Randomized crossover study of the antiemetic activity of levonantradol and metoclopramide in cancer patients receiving chemotherapy.  Cancer Chemother Pharmacol. 1984;13(2):123-125.PubMedGoogle ScholarCrossref
34.
Hutcheon  AW, Palmer  JB, Soukop  M,  et al.  A randomised multicentre single blind comparison of a cannabinoid anti-emetic (levonantradol) with chlorpromazine in patients receiving their first cytotoxic chemotherapy.  Eur J Cancer Clin Oncol. 1983;19(8):1087-1090.PubMedGoogle ScholarCrossref
35.
George  M, Pejovic  MH, Thuaire  M, Kramar  A, Wolff  JP.  [Randomized comparative trial of a new anti-emetic: nabilone, in cancer patients treated with cisplatin].  Biomed Pharmacother. 1983;37(1):24-27.PubMedGoogle Scholar
36.
Jones  SE, Durant  JR, Greco  FA, Robertone  A.  A multi-institutional phase III study of nabilone vs placebo in chemotherapy-induced nausea and vomiting.  Cancer Treat Rev.1982;9(suppl B):45-48.PubMedGoogle ScholarCrossref
37.
Wada  JK, Bogdon  DL, Gunnell  JC, Hum  GJ, Gota  CH, Rieth  TE.  Double-blind, randomized, crossover trial of nabilone vs placebo in cancer chemotherapy.  Cancer Treat Rev.1982;9(suppl B):39-44.PubMedGoogle ScholarCrossref
38.
Johansson  R, Kilkku  P, Groenroos  M. A double-blind, controlled trial of nabilone vs. prochlorperazine for refractory emesis induced by cancer chemotherapy.  Cancer Treat Rev.1982;9(suppl B):25-33.PubMedGoogle ScholarCrossref
39.
Orr  LE, McKernan  JF.  Antiemetic effect of delta 9-tetrahydrocannabinol in chemotherapy-associated nausea and emesis as compared to placebo and compazine.  J Clin Pharmacol.1981;21(8-9 suppl):76S-80S.PubMedGoogle ScholarCrossref
40.
Einhorn  LH, Nagy  C, Furnas  B, Williams  SD.  Nabilone: an effective antiemetic in patients receiving cancer chemotherapy.  J Clin Pharmacol.1981;21(8-9 suppl):64S-69S.PubMedGoogle ScholarCrossref
41.
Orr  LE, McKernan  JF, Bloome  B.  Antiemetic effect of tetrahydrocannabinol: compared with placebo and prochlorperazine in chemotherapy-associated nausea and emesis.  Arch Intern Med. 1980;140(11):1431-1433.PubMedGoogle ScholarCrossref
42.
Steele  N, Gralla  RJ, Braun  DW  Jr, Young  CW.  Double-blind comparison of the antiemetic effects of nabilone and prochlorperazine on chemotherapy-induced emesis.  Cancer Treat Rep. 1980;64(2-3):219-224.PubMedGoogle Scholar
43.
Sallan  SE, Cronin  C, Zelen  M, Zinberg  NE.  Antiemetics in patients receiving chemotherapy for cancer: a randomized comparison of delta-9-tetrahydrocannabinol and prochlorperazine.  N Engl J Med. 1980;302(3):135-138.PubMedGoogle ScholarCrossref
44.
Frytak  S, Moertel  CG, O’Fallon  JR,  et al.  Delta-9-tetrahydrocannabinol as an antiemetic for patients receiving cancer chemotherapy: a comparison with prochlorperazine and a placebo.  Ann Intern Med. 1979;91(6):825-830.PubMedGoogle ScholarCrossref
45.
Ahmedzai  S, Carlyle  DL, Calder  IT, Moran  F.  Anti-emetic efficacy and toxicity of nabilone, a synthetic cannabinoid, in lung cancer chemotherapy.  Br J Cancer. 1983;48(5):657-663.PubMedGoogle ScholarCrossref
46.
Ungerleider  JT, Andrysiak  T, Fairbanks  L, Goodnight  J, Sarna  G, Jamison  K.  Cannabis and cancer chemotherapy: a comparison of oral delta-9-THC and prochlorperazine.  Cancer. 1982;50(4):636-645.PubMedGoogle ScholarCrossref
47.
Sheidler  VR, Ettinger  DS, Diasio  RB, Enterline  JP, Brown  MD.  Double-blind multiple-dose crossover study of the antiemetic effect of intramuscular levonantradol compared to prochlorperazine.  J Clin Pharmacol. 1984;24(4):155-159.PubMedGoogle ScholarCrossref
48.
Harden-Harrison  MM, Munsell  MF, Fisch  MJ,  et al.  Dronabinol for the prevention of nausea from cyclophosphamide and/or adriamycin. Paper presented at: International MASCC/ISOO Symposium: Supportive Care in Cancer; June 28-30, 2012; New York, NY. Support Care Cancer. 2012;20:S209-S210.
49.
Grunberg  SM, Munsell  MF, Morrow  PKH,  et al.  Randomized double-blind evaluation of dronabinol for the prevention of chemotherapy-induced nausea. Paper presented at: Annual Meeting of the American Society of Clinical Oncology (ASCO); 1-5 Jun 2012; Chicago, IL. J Clin Oncol. 2012;30(15)(suppl 1):9061.
50.
Lane  M, Vogel  CL, Ferguson  J,  et al.  Dronabinol and prochlorperazine in combination are better than either single agent alone for treatment of chemotherapy-induced nausea and vomiting.  Proc Am Soc Clin Oncol. 1989;8:326.Google Scholar
51.
Levitt  M. Nabilone vs placebo in the treatment of chemotherapy-induced nausea and vomiting in cancer patients.  Cancer Treat Rev.1982;9(suppl B):49-53.PubMedGoogle ScholarCrossref
52.
Chan  HS, MacLeod  SM, Correia  JA.  Nabilone vs prochlorperazine for control of cancer chemotherapy-induced emesis in children.  Proc Am Soc Clin Oncol. 1984;3:108.Google Scholar
53.
Solvay Pharmaceuticals.  Dronabinol versus standard ondansetron antiemetic therapy in preventing delayed-onset chemotherapy-induced nausea and vomiting. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00642512 Accessed April 7, 2014.
54.
Frytak  S, Moertel  CG, Ofallon  JR.  Comparison of delta-9-tetrahydrocannabinol (THC), prochlorperazine (PCP) and placebo as anti-emetics for cancer-chemotherapy.  Proc Am Assoc Cancer Res. 1979;20:391.Google Scholar
55.
Jhangiani  H, Vredenburgh  JJ, Barbato  L,  et al.  Dronabinol or ondansetron alone and combined for delayed chemotherapy-induced nausea and vomiting (CINV).  Blood. 2005;106(11, part 2):477B.PubMedGoogle Scholar
56.
McCabe  M, Smith  FP, Goldberg  D,  et al.  Comparative trial of oral 9 tetrahydrocannabinol and prochlorperazine for cancer chemotherapy related nausea and vomiting.  Proc Am Assoc Cancer Res and Am Soc Clin Oncol.1981;22:416.Google Scholar
57.
Herman  TS, Einhorn  LH, Jones  SE,  et al.  Superiority of nabilone over prochlorperazine as an antiemetic in patients receiving cancer chemotherapy.  N Engl J Med. 1979;300(23):1295-1297.PubMedGoogle ScholarCrossref
58.
Melhem-Bertrandt  AI, Munsell  MF, Fisch  MJ,  et al A randomized, double-blind, placebo-controlled trial of palonosetron plus dexamethasone with or without dronabinol for the prevention of chemotherapy-induced nausea and vomiting after moderately emetogenic chemotherapy [Unpublished manuscript].2014:1-23.
59.
Abrams  DI, Hilton  JF, Leiser  RJ,  et al.  Short-term effects of cannabinoids in patients with HIV-1 infection: a randomized, placebo-controlled clinical trial.  Ann Intern Med. 2003;139(4):258-266.PubMedGoogle ScholarCrossref
60.
Timpone  JG, Wright  DJ, Li  N,  et al; Division of AIDS Treatment Research Initiative.  The safety and pharmacokinetics of single-agent and combination therapy with megestrol acetate and dronabinol for the treatment of HIV wasting syndrome: the DATRI 004 Study Group.  AIDS Res Hum Retroviruses. 1997;13(4):305-315.PubMedGoogle ScholarCrossref
61.
Struwe  M, Kaempfer  SH, Geiger  CJ,  et al.  Effect of dronabinol on nutritional status in HIV infection.  Ann Pharmacother. 1993;27(7-8):827-831.PubMedGoogle Scholar
62.
Beal  JE, Olson  R, Laubenstein  L,  et al.  Dronabinol as a treatment for anorexia associated with weight loss in patients with AIDS.  J Pain Symptom Manage. 1995;10(2):89-97.PubMedGoogle ScholarCrossref
63.
Ware  M, Fitzcharles  MA, Joseph  L, Shir  Y.  The effects of nabilone on sleep in fibromyalgia: results of a randomized controlled trial. Paper presented at: Canadian Rheumatology Association Meeting; February 18-21, 2009; Kananaskis, AB: Canada. Abstract 149  J Rheumatol. 2009;36(11):2607.Google Scholar
64.
Ware  MA, Fitzcharles  M-A, Joseph  L, Shir  Y.  The effects of nabilone on sleep in fibromyalgia: results of a randomized controlled trial.  Anesth Analg. 2010;110(2):604-610.PubMedGoogle ScholarCrossref
65.
Langford  RM, Mares  J, Novotna  A,  et al.  A double-blind, randomized, placebo-controlled, parallel-group study of THC/CBD oromucosal spray in combination with the existing treatment regimen, in the relief of central neuropathic pain in patients with multiple sclerosis.  J Neurol. 2013;260(4):984-997.PubMedGoogle ScholarCrossref
66.
Wilsey  B, Marcotte  T, Deutsch  R, Gouaux  B, Sakai  S, Donaghe  H.  Low-dose vaporized cannabis significantly improves neuropathic pain.  J Pain. 2013;14(2):136-148.PubMedGoogle ScholarCrossref
67.
Portenoy  RK, Ganae-Motan  ED, Allende  S,  et al.  Nabiximols for opioid-treated cancer patients with poorly-controlled chronic pain: a randomized, placebo-controlled, graded-dose trial.  J Pain. 2012;13(5):438-449.PubMedGoogle ScholarCrossref
68.
Ware  MA, Wang  T, Shapiro  S,  et al.  Smoked cannabis for chronic neuropathic pain: a randomized controlled trial.  CMAJ. 2010;182(14):E694-E701.PubMedGoogle ScholarCrossref
69.
Johnson  JR, Burnell-Nugent  M, Lossignol  D, Ganae-Motan  ED, Potts  R, Fallon  MT.  Multicenter, double-blind, randomized, placebo-controlled, parallel-group study of the efficacy, safety, and tolerability of THC:CBD extract and THC extract in patients with intractable cancer-related pain.  J Pain Symptom Manage. 2010;39(2):167-179.PubMedGoogle ScholarCrossref
70.
Selvarajah  D, Gandhi  R, Emery  CJ, Tesfaye  S.  Randomized placebo-controlled double-blind clinical trial of cannabis-based medicinal product (Sativex) in painful diabetic neuropathy: depression is a major confounding factor.  Diabetes Care. 2010;33(1):128-130.PubMedGoogle ScholarCrossref
71.
Ellis  RJ, Toperoff  W, Vaida  F,  et al.  Smoked medicinal cannabis for neuropathic pain in HIV: a randomized, crossover clinical trial.  Neuropsychopharmacology. 2009;34(3):672-680.PubMedGoogle ScholarCrossref
72.
Wilsey  B, Marcotte  T, Tsodikov  A,  et al.  A randomized, placebo-controlled, crossover trial of cannabis cigarettes in neuropathic pain.  J Pain. 2008;9(6):506-521.PubMedGoogle ScholarCrossref
73.
Narang  S, Gibson  D, Wasan  AD,  et al.  Efficacy of dronabinol as an adjuvant treatment for chronic pain patients on opioid therapy.  J Pain. 2008;9(3):254-264.PubMedGoogle ScholarCrossref
74.
Skrabek  RQ, Galimova  L, Ethans  K, Perry  D.  Nabilone for the treatment of pain in fibromyalgia.  J Pain. 2008;9(2):164-173.PubMedGoogle ScholarCrossref
75.
Frank  B, Serpell  MG, Hughes  J, Matthews  JNS, Kapur  D.  Comparison of analgesic effects and patient tolerability of nabilone and dihydrocodeine for chronic neuropathic pain: randomised, crossover, double blind study.  BMJ. 2008;336(7637):199-201.PubMedGoogle ScholarCrossref
76.
Nurmikko  TJ, Serpell  MG, Hoggart  B, Toomey  PJ, Morlion  BJ, Haines  D.  Sativex successfully treats neuropathic pain characterised by allodynia: a randomised, double-blind, placebo-controlled clinical trial.  Pain. 2007;133(1-3):210-220.PubMedGoogle ScholarCrossref
77.
Abrams  DI, Jay  CA, Shade  SB,  et al.  Cannabis in painful HIV-associated sensory neuropathy: a randomized placebo-controlled trial.  Neurology. 2007;68(7):515-521.PubMedGoogle ScholarCrossref
78.
Pinsger  M, Schimetta  W, Volc  D, Hiermann  E, Riederer  F, Pölz  W.  [Benefits of an add-on treatment with the synthetic cannabinomimetic nabilone on patients with chronic pain—a randomized controlled trial].  Wien Klin Wochenschr. 2006;118(11-12):327-335.PubMedGoogle ScholarCrossref
79.
Blake  DR, Robson  P, Ho  M, Jubb  RW, McCabe  CS.  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). 2006;45(1):50-52.PubMedGoogle ScholarCrossref
80.
Rog  DJ, Nurmikko  TJ, Friede  T, Young  CA.  Randomized, controlled trial of cannabis-based medicine in central pain in multiple sclerosis.  Neurology. 2005;65(6):812-819.PubMedGoogle ScholarCrossref
81.
Berman  JS, Symonds  C, Birch  R.  Efficacy of two cannabis based medicinal extracts for relief of central neuropathic pain from brachial plexus avulsion: results of a randomised controlled trial.  Pain. 2004;112(3):299-306.PubMedGoogle ScholarCrossref
82.
Svendsen  KB, Jensen  TS, Bach  FW.  Does the cannabinoid dronabinol reduce central pain in multiple sclerosis? randomised double blind placebo controlled crossover trial.  BMJ. 2004;329(7460):253.PubMedGoogle ScholarCrossref
83.
Karst  M, Salim  K, Burstein  S, Conrad  I, Hoy  L, Schneider  U.  Analgesic effect of the synthetic cannabinoid CT-3 on chronic neuropathic pain: a randomized controlled trial.  JAMA. 2003;290(13):1757-1762.PubMedGoogle ScholarCrossref
84.
Noyes  R  Jr, Brunk  SF, Baram  DA, Canter  A.  Analgesic effect of delta-9-tetrahydrocannabinol.  J Clin Pharmacol. 1975;15(2-3):139-143.PubMedGoogle ScholarCrossref
85.
Lynch  ME, Cesar-Rittenberg  P, Hohmann  AG.  A double-blind, placebo-controlled, crossover pilot trial with extension using an oral mucosal cannabinoid extract for treatment of chemotherapy-induced neuropathic pain.  J Pain Symptom Manage. 2014;47(1):166-173.PubMedGoogle ScholarCrossref
86.
Wallace  M, Atkinson  J, Gouaux  B, Marcotte  T, Umlauf  A.  Effect of smoked cannabis on painful diabetic peripheral neuropathy. Paper presented at: 32nd Annual Scientific Meeting of the American Pain Society; May 9-11, 2013; New Orleans: LA.  J Pain. 2013;14(4)(suppl 1):S62 doi:10.1016/j.jpain.2013.01.587.Google ScholarCrossref
87.
Berman  J, Bosworth  T, Guy  G, Stott  C; Sativex Spinal Cord Injury Study Group.  Sativex in the treatment of central neuropathic pain due to spinal cord injury: a randomised controlled study. Paper presented at: British Pain Society Annual Scientific Meeting; April 2007; Glasgow: United Kingdom.
88.
Serpell  M, Ratcliffe  S, Hovorka  J,  et al.  A double-blind, randomized, placebo-controlled, parallel group study of THC/CBD spray in peripheral neuropathic pain treatment.  Eur J Pain. 2014;18(7):999-1012.PubMedGoogle ScholarCrossref
89.
Fitzcharles  MA, Shir  Y, Joseph  L, Ware  MA.  The effects of nabilone on insomnia in fibromyalgia: results of a randomized controlled trial. Paper presented at: American College of Rheumatology/Association of Rheumatology Health Professionals Annual Scientific Meeting (ACR/ARHP 09); November 6-11, 2009; Atlanta: GA.  Arthritis Rheum. 2009;60:1429.Google Scholar
90.
McGill University Health Center.  Nabilone versus amitriptyline in improving quality of sleep in patients with fibromyalgia. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00381199 Accessed April 7, 2014.
91.
GW Pharmaceuticals Ltd.  Sativex versus placebo when added to existing treatment for central neuropathic pain in MS.http://ClinicalTrials.gov/show/NCT00391079 Accessed April 7, 2014.
92.
Svendsen  KB, Jensen  TS, Bach  FW.  [Effect of the synthetic cannabinoid dronabinol on central pain in patients with multiple sclerosis–secondary publication].  Ugeskr Laeger.2005;167(25-31):2772-2774.PubMedGoogle Scholar
93.
Salim  K, Schneider  U, Burstein  S, Hoy  L, Karst  M.  Pain measurements and side effect profile of the novel cannabinoid ajulemic acid.  Neuropharmacology. 2005;48(8):1164-1171.PubMedGoogle ScholarCrossref
94.
Pinsger  M.  Benefit of an add-on-treatment with a synthetic cannabinomimeticum on patients with chronic back pain-a randomized controlled trial. Paper presented at 8th International Conference on Early Psychosis: From Neurobiology to Public Policy; October 11-13, 2012; San Francisco: CA.  Eur Spine J. 2012;21(11):2366 doi:10.1007/s00586-012-2522-6.Google Scholar
95.
Nurmikko  TJ, Serpell  MG, Hoggart  B, Toomey  PJ, Morlion  BJ.  A multi-centre, double-blind, randomized, controlled trial of oro-mucosal cannabis based medicine in the treatment of neuropathic pain characterized by allodynia.  Neurology. 2005;64(suppl 1):A374.Google ScholarCrossref
96.
Issa  MA, Narang  S, Jamison  RN,  et al.  The subjective psychoactive effects of oral dronabinol studied in a randomized, controlled crossover clinical trial for pain.  Clin J Pain. 2014;30(6):472-478.PubMedGoogle ScholarCrossref
97.
Abrams  DI, Jay  CA, Vizoso  H,  et al.  Smoked cannabis therapy for HIV-related painful peripheral neuropathy: results of a randomized, placebo-controlled clinical trial. Paper presented at: IACM 3rd Conference on Cannabinoids in Medicine; September 9-10, 2005; Leiden, the Netherlands.
98.
Young  CA, Rog  DJ.  Randomised controlled trial of cannabis based medicinal extracts (CBME) in central neuropathic pain due to multiple sclerosis. Paper presented at: IV Congress of the European Federation of IASP Chapters (EFIC); September 2-6, 2003; Prague, Czech Republic.
99.
Berman  J, Lee  J, Cooper  M,  et al.  Efficacy of two cannabis-based medicinal extracts for relief of central neuropathic pain from brachial plexus avulsion: results of a randomised controlled trial. Paper presented at: Pain Society Annual Meeting; April 1-4, 2003; Glasgow, United Kingdom.  Anaesthesia. 2003;58(9):938 doi:10.1046/j.1365-2044.2003.03408_3.x.Google ScholarCrossref
100.
Center for Medicinal Cannabis Research.  Effects of smoked marijuana on neuropathic pain. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00254761. Accessed April 7, 2014.
101.
Center for Medicinal Cannabis Research.  Medicinal cannabis for painful HIV neuropathy. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00255580. Accessed April 7, 2014.
102.
University of California Davis.  Center for Medicinal Cannabis Research, VA Northern California Health Care System. Effects of vaporized marijuana on neuropathic pain. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT01037088. Accessed April 7, 2014.
103.
Center for Medicinal Cannabis Research.  Marijuana for HIV-related peripheral neuropathy. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00046722. Accessed April 7, 2014.
104.
GW Pharmaceuticals Ltd.  A study of Sativex® for pain relief in patients with advanced malignancy. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00530764. Accessed April 7, 2014.
105.
GW Pharmaceuticals Ltd.  A study of sativex® for pain relief in patients with advanced malignancy. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00674609. Accessed April 7, 2014.
106.
GW Pharmaceuticals Ltd.  A study of sativex® for relief of peripheral neuropathic pain associated with allodynia. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00711880. Accessed April 7, 2014.
107.
GW Pharmaceuticals Ltd.  A study of sativex in the treatment of central neuropathic pain due to multiple sclerosis. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT01604265. Accessed April 7, 2014.
108.
GW Pharmaceuticals Ltd.  A study of sativex® for pain relief due to diabetic neuropathy. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00710424. Accessed April 7, 2014.
109.
GW Pharmaceuticals Ltd.  A study of Sativex® for pain relief of peripheral neuropathic pain, associated with allodynia. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00710554. Accessed April 7, 2014.
110.
Mary Lynch, Capital District Health Authority Canada.  Sativex for treatment of chemotherapy induced neuropathic pain. .ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00872144. Accessed April 7, 2014.
111.
Brigham and Women’s Hospital; Solvay Pharmaceuticals.  Study to evaluate the efficacy of dronabinol (Marinol) as add-on therapy for patients on opioids for chronic pain. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00153192. Accessed April 7, 2014.
112.
Winnipeg Regional Health Authority; Valeant Canada Limited.  A trial assessing the effect of nabilone on pain and quality of life in patients with fibromyalgia. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00272207. Accessed April 7, 2014.
113.
GW Pharma Ltd.  A double blind, randomised, placebo controlled parallel group study of cannabis based medicine extract (CBME), in the treatment of peripheral neuropathic pain characterised by allodynia. metaRegister of Controlled Trials. http://www.controlled-trials.com/ISRCTN38250575. Accessed April 7, 2014.
114.
Montreal General Hospital.  Pilot study of smoked cannabis for chronic neuropathic pain. metaRegister of Controlled Trials (mRCT), http://www.controlled-trials.com/ISRCTN68314063. Accessed April 7, 2014.
115.
GW Pharma Ltd.  A double blind, randomised, placebo controlled, parallel group study of Sativex, in the treatment of subjects with peripheral neuropathic pain associated with allodynia. EU Clinical Trials Register. https://www.clinicaltrialsregister.eu/ctr-search/search?query=eudract_number:2004-002531-32. Accessed April 8, 2014.
116.
Cambridge Laboratories Ltd.  A randomised, crossover, double blind comparison of the analgesic effect and patient tolerability of nabilone and dihydrocodeine in chronic neuropathic pain. metaRegister of Controlled Trials. http://isrctn.org/ISRCTN15330757. Accessed April 7, 2014.
117.
Selvarajah  D, Gandhi  RA, Witte  D, Bowler  H, Emery  C, Tesfaye  S.  Treatment of painful diabetic neuropathy with Sativex (a cannabis based medicinal product)—results of a randomised placebo controlled trial.  Diabetologia. 2006;49 (suppl 1):671-672 doi:10.1007/s00125-006-0358-5.Google Scholar
118.
Rog  DJ, Nurmikko  T, Young  C, Sarantis  NS.  Randomized controlled trial of sativex, a cannabis based medicine (CBM), in central neuropathic pain due to multiple sclerosis, followed by an open-label extension.  Neurology. 2006;66(5):A31.Google Scholar
119.
Ventegodt  S, Merrick  J.  Psychoactive drugs and quality of life.  ScientificWorldJournal. 2003;3:694-706.PubMedGoogle ScholarCrossref
120.
Wallace  MS, Marcotte  TD, Umlauf  A, Gouaux  B, Atkinson  JH.  Efficacy of inhaled cannabis on painful diabetic neuropathy. J Pain.2015;pii:S1526-5900(1515)00601-X.PubMed
121.
Turcotte  D, Doupe  M, Torabi  M,  et al.  Nabilone as an adjunctive to gabapentin for multiple sclerosis-induced neuropathic pain: a randomized controlled trial.  Pain Med. 2015;16(1):149-159.PubMedGoogle ScholarCrossref
122.
Zajicek  JP, Sanders  HP, Wright  DE,  et al.  Cannabinoids in multiple sclerosis (CAMS) study: safety and efficacy data for 12 months follow up.  J Neurol Neurosurg Psychiatry. 2005;76(12):1664-1669.PubMedGoogle ScholarCrossref
123.
Zajicek  JP, Hobart  JC, Slade  A, Barnes  D, Mattison  PG; MUSEC Research Group.  Multiple sclerosis and extract of cannabis: results of the MUSEC trial.  J Neurol Neurosurg Psychiatry. 2012;83(11):1125-1132.PubMedGoogle ScholarCrossref
124.
Corey-Bloom  J, Wolfson  T, Gamst  A,  et al.  Smoked cannabis for spasticity in multiple sclerosis: a randomized, placebo-controlled trial.  CMAJ. 2012;184(10):1143-1150.PubMedGoogle ScholarCrossref
125.
Collin  C, Ehler  E, Waberzinek  G,  et al.  A double-blind, randomized, placebo-controlled, parallel-group study of Sativex, in subjects with symptoms of spasticity due to multiple sclerosis.  Neurol Res. 2010;32(5):451-459.PubMedGoogle ScholarCrossref
126.
Pooyania  S, Ethans  K, Szturm  T, Casey  A, Perry  D.  A randomized, double-blinded, crossover pilot study assessing the effect of nabilone on spasticity in persons with spinal cord injury.  Arch Phys Med Rehabil. 2010;91(5):703-707.PubMedGoogle ScholarCrossref
127.
Collin  C, Davies  P, Mutiboko  IK, Ratcliffe  S; Sativex Spasticity in MS Study Group.  Randomized controlled trial of cannabis-based medicine in spasticity caused by multiple sclerosis.  Eur J Neurol. 2007;14(3):290-296.PubMedGoogle ScholarCrossref
128.
Freeman  RM, Adekanmi  O, Waterfield  MR, Waterfield  AE, Wright  D, Zajicek  J.  The effect of cannabis on urge incontinence in patients with multiple sclerosis: a multicentre, randomised placebo-controlled trial (CAMS-LUTS).  Int Urogynecol J Pelvic Floor Dysfunct. 2006;17(6):636-641.PubMedGoogle ScholarCrossref
129.
Wade  DT, Makela  P, Robson  P, House  H, Bateman  C.  Do cannabis-based medicinal extracts have general or specific effects on symptoms in multiple sclerosis? a double-blind, randomized, placebo-controlled study on 160 patients.  Mult Scler. 2004;10(4):434-441.PubMedGoogle ScholarCrossref
130.
Vaney  C, Heinzel-Gutenbrunner  M, Jobin  P,  et al.  Efficacy, safety and tolerability of an orally administered cannabis extract in the treatment of spasticity in patients with multiple sclerosis: a randomized, double-blind, placebo-controlled, crossover study.  Mult Scler. 2004;10(4):417-424.PubMedGoogle ScholarCrossref
131.
Zajicek  J, Fox  P, Sanders  H,  et al; UK MS Research Group.  Cannabinoids for treatment of spasticity and other symptoms related to multiple sclerosis (CAMS study): multicentre randomised placebo-controlled trial.  Lancet. 2003;362(9395):1517-1526.PubMedGoogle ScholarCrossref
132.
Killestein  J, Hoogervorst  ELJ, Reif  M,  et al.  Safety, tolerability, and efficacy of orally administered cannabinoids in MS.  Neurology. 2002;58(9):1404-1407.PubMedGoogle ScholarCrossref
133.
Hobart  JC, Zajicek  JP.  Cannabis as a symptomatic treatment for MS: Clinically meaningful MUSEC to the stiffness and walking problems of people with MS. Paper presented at: 28th Congress of the European Committee for Treatment and Research in Multiple Sclerosis; October 10-13, 2012; Lyon: France.  Mult Scler. 2012;18(4 suppl 1):247.PubMedGoogle Scholar
134.
Zajicek  J, Reif  M, Schnelle  M.  Cannabis extract in the treatment of muscle stiffness and other symptoms in multiple sclerosis—Results of the MUSEC study. Paper presented at: 25th Congress of the European Committee for Treatment and Research in Multiple Sclerosis; September 9-12, 2009; Dusseldorf: Germany.  Mult Scler. 2009;15(9)(suppl S):S274 doi:10.1177/1352458509107025.Google Scholar
135.
Killestein  J, Hoogervorst  ELJ, Kalkers  NF,  et al.  The effects of orally administred cannabinoids in multiple sclerosis patients: a pilot study.  Mult Scler. 2000;6(1 suppl 1):S28 doi:10.1177/135245850000600101.Google Scholar
136.
Zajicek  J, Reif  M, Schnelle  M; UK MUSEC Study Investigators.  Cannabis extract in the treatment of muscle stiffness and other symptoms in multiple sclerosis – results of the MUSEC study. Paper presented at: IACM 5th Conference on Cannabinoids in Medicine; October 2-3, 2009; Cologne, Germany.
137.
Collin  C, Ambler  Z, Kent  R, McCalla  R.  A randomised controlled study of Sativex® in patients with symptoms of spasticity due to multiple sclerosis. Paper presented at: 22nd Congress of the ECTRIMS; September 27-30, 2006; Madrid, Spain.
138.
Robson  P, Wade  D, Makela  P, House  H, Bateman  C.  Cannabis-based medicinal extract (Sativex) produced significant improvements in a subjective measure of spasticity which were maintained on long-term treatment with no evidence of tolerance. Paper presented at: IACM 3rd Conference on Cannabinoids in Medicine; September 9-10, 2005; Leiden, the Netherlands.
139.
Center for Medicinal Cannabis Research.  Short-term effects of medicinal cannabis therapy on spasticity in multiple sclerosis. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00248378. Accessed April 7, 2014.
140.
Institut fur Klinische Forschung Germany; Weleda AG. Multiple Sclerosis and Extract of Cannabis (MUSEC) study. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00552604. Accessed April 7, 2014.
141.
GW Pharmaceuticals Ltd.  A study of Sativex® for relief of spasticity in subjects with multiple sclerosis. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00711646. Accessed April 7, 2014.
142.
GW Pharmaceuticals Ltd.  A study to evaluate the efficacy of Sativex in relieving symptoms of spasticity due to multiple sclerosis. ClinicalTrials.gov.http://ClinicalTrials.gov/show/NCT01599234. Accessed April 7, 2014.
143.
GW Pharmaceuticals Ltd.  An investigation of delta-9-tetrahydrocannabinol (THC) and cannabidiol (CBD) in multiple sclerosis patients. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT01610700. Accessed April 7, 2014.
144.
University of Manitoba, Valeant Canada Limited.  Randomized double blind cross over study for nabilone in spasticity in spinal cord injury persons. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00623376. Accessed April 7, 2014.
145.
Medical Research Council (MRC).  A multiple randomised controlled trial of cannabinoids on spasticity in multiple sclerosis (MS). metaRegister of Controlled Trials. http://www.controlled-trials.com/ISRCTN39371386. Accessed April 7, 2014.
146.
Gesellschaft fuer klinische Forschung e.V.  Multiple Sclerosis and Extract of Cannabis (MUSEC): a randomised, double-blind, placebo-controlled phase III trial to determine the efficacy and safety of a standardised oral extract of cannabis sativa for the symptomatic relief of muscle stiffness and pain in multiple sclerosis. EU Clinical Trials Register. https://www.clinicaltrialsregister.eu/ctr-search/search?query=eudract_number:2005-005263-29. Accessed April 8, 2014.
147.
Corey-Bloom  J, Wolfson  TJ, Anthony  GC, Bentley  H, Gouaux  B.  Short-term effects off medicinal cannabis on spasticity in multiple sclerosis.  Neurology. 2008;70(11)(suppl 1):A86-A87.Google Scholar
148.
Leocani  L, Nuara  A, Houdayer  E,  et al.  Effect of THC-CBD oromucosal spray (Sativex) on measures of spasticity in multiple sclerosis: a double-blind, placebo-controlled, crossover study. Paper presented at: Joint Americas Committee for Treatment and Research in Multiple Sclerosis ACTRIMS—European Committee for Treatment and Research in Multiple Sclerosis ECTRIMS Meeting; September 10-13, 2014; Boston, MA.  Mult Scler. 2014;20(1 suppl 1):498 doi:10.1177/1352458514547846.Google Scholar
149.
Van Amerongen  G, Beumer  T, Killestein  J, Groeneveld  GJ.  Individualized dosing of a novel oral DELTA9-THC formulation improves subjective spasticity and pain in patients with progressive multiple sclerosis. Paper presented at Joint Americas Committee for Treatment and Research in Multiple Sclerosis ACTRIMS—European Committee for Treatment and Research in Multiple Sclerosis ECTRIMS Meeting; September 10-13, 2014; Boston,MA.  Mult Scler. 2014;20(1)(suppl 1):478-479 doi:10.1177/1352458514547846.Google Scholar
150.
Bergamaschi  MM, Queiroz  RH, Chagas  MH,  et al.  Cannabidiol reduces the anxiety induced by simulated public speaking in treatment-naïve social phobia patients.  Neuropsychopharmacology. 2011;36(6):1219-1226.PubMedGoogle ScholarCrossref
151.
Leweke  FM, Piomelli  D, Pahlisch  F,  et al.  Cannabidiol enhances anandamide signaling and alleviates psychotic symptoms of schizophrenia.  Transl Psychiatry. 2012;2:e94.PubMedGoogle ScholarCrossref
152.
Leweke  FM, Gerth  CW, Nolden  BM,  et al.  Cannabidiol as antipsychotic. Paper presented at: 21st ECNP Congress; August 30, 2008; Barcelona, Spain.  Eur Neuropsychopharmacol. 2008;18(S4):S171 doi:10.1016/S0924-977X(08)70156-1.Google ScholarCrossref
153.
Leweke  FM, Koethe  D, Pahlisch  F,  et al.  Antipsychotic effects of cannabidiol. Paper presented at: 17th European Psychiatric Association, EPA Congress; January 24-28, 2009; Lisbon, Portugal.  Eur Psychiatry. 2009;24(supp 1):S207 doi:10.1016/S0924-9338(09)70440-7.Google ScholarCrossref
154.
University of Cologne.  Evaluation of the antipsychotic efficacy of cannabidiol in acute schizophrenic psychosis. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00628290. Accessed April 7, 2014.
155.
Rohleder  C, Pahlisch  F, Schaefer  C,  et al.  The endocannabinoid system as a pharmacological target for antipsychotic treatment and more? Paper presented at: 8th International Conference on Early Psychosis: From Neurobiology to Public Policy; October 11-13, 2012; San Francisco, CA.  Early Interv Psychiatry. 2012;6(suppl 1):7 doi:10.1111/j.1751-7893.2012.00392.x.Google Scholar
156.
Markus  F, Leweke  M, Kranaster  L,  et al.  The efficacy of cannabidiol in the treatment of schizophrenia—A translational approach. Paper presented at: 13th International Congress on Schizophrenia Research, ICOSR; April 2-6, 2011; Colorado Springs, CO.  Schizophr Bull. 2011;37(suppl 1):313 doi:10.1093/schbul/sbq173.Google Scholar
157.
Leweke  FM, Kranaster  L, Hellmich  M, Koethe  D.  Cannabidiol as a new type of an antipsychotic: results from a placebo-controlled clinical trial. Paper presented at: 49th Annual Conference of the American College of Neuropsychopharmacology, ACNP 2010; December 5-9, 2010; Miami Beach, FL.  Neuropsychopharmacology. 2010;35(suppl 1):S280 doi:10.1038/npp.2010.217.Google Scholar
158.
Leweke  FM, Hellmich  M, Kranaster  L, Koethe  D.  Cannabidiol as a new type of an antipsychotic: results from a placebo-controlled clinical trial. Paper presented at: 67th Annual Scientific Convention and Meeting of the Society of Biological Psychiatry; May 3-5, 2012; Philadelphia, PA. Biol Psychiatry. 2012;78(8)(suppl 1):63S.
159.
Tomida  I, Azuara-Blanco  A, House  H, Flint  M, Pertwee  RG, Robson  PJ.  Effect of sublingual application of cannabinoids on intraocular pressure: a pilot study.  J Glaucoma. 2006;15(5):349-353.PubMedGoogle ScholarCrossref
160.
Müller-Vahl  KR, Prevedel  H, Theloe  K, Kolbe  H, Emrich  HM, Schneider  U.  Treatment of Tourette syndrome with delta-9-tetrahydrocannabinol (delta 9-THC): no influence on neuropsychological performance.  Neuropsychopharmacology. 2003;28(2):384-388.PubMedGoogle ScholarCrossref
161.
Müller-Vahl  KR, Schneider  U, Prevedel  H,  et al.  Delta 9-tetrahydrocannabinol (THC) is effective in the treatment of tics in Tourette syndrome: a 6-week randomized trial.  J Clin Psychiatry. 2003;64(4):459-465.PubMedGoogle ScholarCrossref
162.
Müller-Vahl  KR, Koblenz  A, Jöbges  M, Kolbe  H, Emrich  HM, Schneider  U.  Influence of treatment of Tourette syndrome with delta9-tetrahydrocannabinol (delta9-THC) on neuropsychological performance.  Pharmacopsychiatry. 2001;34(1):19-24.PubMedGoogle ScholarCrossref
163.
Müller-Vahl  KR, Schneider  U, Koblenz  A,  et al.  Treatment of Tourette’s syndrome with Delta 9-tetrahydrocannabinol (THC): a randomized crossover trial.  Pharmacopsychiatry. 2002;35(2):57-61.PubMedGoogle ScholarCrossref
164.
International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use.  MeDRA (Medical Dictionary for Regulatory Activities).http://www.meddra.org/. Accessed September 2, 2014.
165.
McGowan  J, Sampson  M, Lefebvre  C.  An evidence based checklist for the peer review of electronic search strategies (PRESS EBC).  Evid Based Libr Inf Pract. 2010;5(1):1-6. http://ejournals.library.ualberta.ca/index.php/EBLIP/article/view/7402. Accessed Month, date, year.Google Scholar
166.
Elbourne  DR, Altman  DG, Higgins  JP, Curtin  F, Worthington  HV, Vail  A.  Meta-analyses involving cross-over trials: methodological issues.  Int J Epidemiol. 2002;31(1):140-149.PubMedGoogle ScholarCrossref
167.
Lakhan  SE, Rowland  M.  Whole plant cannabis extracts in the treatment of spasticity in multiple sclerosis: a systematic review.  BMC Neurol. 2009;9:59.PubMedGoogle ScholarCrossref
168.
Sevilla Guerra  S.  Are cannabinoids more effective than placebo in decreasing MS-related bladder dysfunction?  Br J Neurosci Nurs. 2012;8(2):71-78 doi:10.12968/bjnn.2012.8.2.71.Google ScholarCrossref
169.
Shakespeare  D, Boggild  M, Young  CA.  Anti-spasticity agents for multiple sclerosis.  Cochrane Database Syst Rev. 2003;(4):CD001332.PubMedGoogle Scholar
170.
Wade  DT, Collin  C, Stott  C, Duncombe  P.  Meta-analysis of the efficacy and safety of Sativex (nabiximols), on spasticity in people with multiple sclerosis.  Mult Scler. 2010;16(6):707-714.PubMedGoogle ScholarCrossref
171.
Brook  JS, Lee  JY, Finch  SJ, Brown  EN.  Course of comorbidity of tobacco and marijuana use: psychosocial risk factors.  Nicotine Tob Res. 2010;12(5):474-482.PubMedGoogle ScholarCrossref
172.
Machado Rocha  FC, Stéfano  SC, De Cássia Haiek  R, Rosa Oliveira  LM, Da Silveira  DX.  Therapeutic use of Cannabis sativa on chemotherapy-induced nausea and vomiting among cancer patients: systematic review and meta-analysis.  Eur J Cancer Care (Engl). 2008;17(5):431-443.PubMedGoogle ScholarCrossref
173.
Phillips  RS, Gopaul  S, Gibson  F,  et al Antiemetic medication for prevention and treatment of chemotherapy induced nausea and vomiting in childhood.  Cochrane Database Syst Rev. 2010;(9):CD007786.PubMedGoogle Scholar
174.
Tramèr  MR, Carroll  D, Campbell  FA, Reynolds  DJ, Moore  RA, McQuay  HJ.  Cannabinoids for control of chemotherapy induced nausea and vomiting: quantitative systematic review.  BMJ. 2001;323(7303):16-21.PubMedGoogle ScholarCrossref
175.
van den Elsen  GAH, Ahmed  AIA, Lammers  M,  et al.  Efficacy and safety of medical cannabinoids in older subjects: a systematic review.  Ageing Res Rev. 2014;14(1):56-64.PubMedGoogle ScholarCrossref
176.
Burns  TL, Ineck  JR.  Cannabinoid analgesia as a potential new therapeutic option in the treatment of chronic pain.  Ann Pharmacother. 2006;40(2):251-260.PubMedGoogle ScholarCrossref
177.
Campbell  FA, Tramèr  MR, Carroll  D, Reynolds  DJ, Moore  RA, McQuay  HJ.  Are cannabinoids an effective and safe treatment option in the management of pain? a qualitative systematic review.  BMJ. 2001;323(7303):13-16.PubMedGoogle ScholarCrossref
178.
Canadian Agency for Drugs and Technologies in Health (CADTH).  Cannabinoids as Co-Analgesics: Review of Clinical Effectiveness. Ottawa, Canada: Canadian Agency for Drugs and Technologies in Health;2010.
179.
Canadian Agency for Drugs and Technologies in Health (CADTH).  Cannabinoids for the Management of Neuropathic Pain: Review of Clinical Effectiveness. Ottawa, Canada: Canadian Agency for Drugs and Technologies in Health);2010.
180.
Davis  MP.  Oral nabilone capsules in the treatment of chemotherapy-induced nausea and vomiting and pain.  Expert Opin Investig Drugs. 2008;17(1):85-95.PubMedGoogle ScholarCrossref
181.
Iskedjian  M, Bereza  B, Gordon  A, Piwko  C, Einarson  TR.  Meta-analysis of cannabis based treatments for neuropathic and multiple sclerosis-related pain.  Curr Med Res Opin. 2007;23(1):17-24.PubMedGoogle ScholarCrossref
182.
Jawahar  R, Oh  U, Yang  S, Lapane  KL.  A systematic review of pharmacological pain management in multiple sclerosis.  Drugs. 2013;73(15):1711-1722.PubMedGoogle ScholarCrossref
183.
Kung  T, Hochman  J, Sun  Y,  et al.  Efficacy and safety of cannabinoids for pain in musculoskeletal diseases: a systematic review and meta-analysis. Paper presented at: 2nd Mexican-Canadian Congress of Rheumatology; February 10-15, 2011; Cancun, Mexico.  J Rheumatol. 2011;38(6):1171 doi:10.3899/jrheum.110506.Google Scholar
184.
Lynch  ME, Campbell  F.  Cannabinoids for treatment of chronic non-cancer pain; a systematic review of randomized trials.  Br J Clin Pharmacol. 2011;72(5):735-744.PubMedGoogle ScholarCrossref
185.
Martín-Sánchez  E, Furukawa  TA, Taylor  J, Martin  JL.  Systematic review and meta-analysis of cannabis treatment for chronic pain.  Pain Med. 2009;10(8):1353-1368.PubMedGoogle ScholarCrossref
186.
Phillips  TJ, Cherry  CL, Cox  S, Marshall  SJ, Rice  AS.  Pharmacological treatment of painful HIV-associated sensory neuropathy: a systematic review and meta-analysis of randomised controlled trials.  PLoS One. 2010;5(12):e14433.PubMedGoogle ScholarCrossref
187.
Pittler  MH, Ernst  E.  Complementary therapies for neuropathic and neuralgic pain: systematic review.  Clin J Pain. 2008;24(8):731-733.PubMedGoogle ScholarCrossref
188.
Richards  BL, Whittle  SL, Buchbinder  R.  Neuromodulators for pain management in rheumatoid arthritis.  Cochrane Database Syst Rev. 2012;(1):CD008921.PubMedGoogle Scholar
189.
Boychuk  DG, Goddard  G, Mauro  G, Orellana  MF.  The effectiveness of cannabinoids in the management of chronic nonmalignant neuropathic pain: a systematic review.  J Oral Facial Pain Headache. 2015;29(1):7-14.PubMedGoogle ScholarCrossref
190.
Parsai  S, Herman  R, Johnson  S.  Systematic literature review of randomized controlled trials to evaluate the efficacy of medical marijuana for analgesia. Paper presented at: Annual Meeting of the American College of Clinical Pharmacy; October 12-15, 2014; Austin, TX.  Pharmacotherapy. 2014;34(10):e287 doi:10.1002/phar.1497.Google Scholar
191.
Lynch  ME, Ware  MA.  Cannabinoids for the treatment of chronic non-cancer pain: an updated systematic review of randomized controlled trials [published online March 22, 2015].  J Neuroimmune Pharmacol doi.org/10.1007/s11481-015-9600-6.PubMedGoogle Scholar
192.
Rathbone  J, Variend  H, Mehta  H. Cannabis and schizophrenia.  Cochrane Database Syst Rev.2008;(3):CD004837.PubMedGoogle Scholar
193.
Schoeler  T, Kambeitz  J, Bhattacharyya  S.  The effect of cannabis on memory function in users with and without a psychotic disorder: a meta-analysis. Paper presented at: 26th European College of Neuropsychopharmacology, ECNP Congress; October 5-9, 2013; Barcelona, Spain.  Eur Neuropsychopharmacol. 2013;23:S216-S217 doi:10.1016/S0924-977X(13)70334-1.Google ScholarCrossref
194.
Zammit  S, Moore  THM, Lingford-Hughes  A,  et al.  Effects of cannabis use on outcomes of psychotic disorders: systematic review.  Br J Psychiatry. 2008;193(5):357-363.PubMedGoogle ScholarCrossref
195.
Curtis  A, Clarke  CE, Rickards  HE.  Cannabinoids for Tourette’s syndrome.  Cochrane Database Syst Rev. 2009;CD0065652009(4):CD006565.PubMedGoogle Scholar
196.
Waldon  K, Hill  J, Termine  C, Balottin  U, Cavanna  AE.  Trials of pharmacological interventions for Tourette syndrome: a systematic review.  Behav Neurol. 2013;26(4):265-273.PubMedGoogle ScholarCrossref
197.
Boers  M.  Updated Consolidated Standards of Reporting Trials (CONSORT): it just gets better.  J Clin Epidemiol. 2010;63(8):813-814.PubMedGoogle ScholarCrossref
Original Investigation
June 23/30, 2015

Cannabinoids for Medical UseA Systematic Review and Meta-analysis

Author Affiliations
  • 1School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
  • 2The National Institute for Health Research Collaboration for Leadership in Applied Health Research and Care West at University Hospitals, Bristol NHS Foundation Trust, Bristol, United Kingdom
  • 3Kleijnen Systematic Reviews Ltd, Escrick, York, United Kingdom
  • 4Department of Medical, Oral, and Biotechnological Sciences, University “G. D'Annunzio” of Chieti-Pescara, Chieti, Italy
  • 5Department of Vascular Medicine, Academic Medical Center, Amsterdam, the Netherlands
  • 6Medical School, Universidad Peruana de Ciencias Aplicadas (UPC), Lima, Peru
  • 7Health Outcomes and Clinical Epidemiology Section, Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
  • 8Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
  • 9Institut für Epidemiologie und kongenitale Erkrankungen, Cepicon GmbH, Hamburg, Germany
  • 10School for Public Health and Primary Care (CAPHRI), Maastricht University, Maastricht, the Netherlands
JAMA. 2015;313(24):2456-2473. doi:10.1001/jama.2015.6358
Abstract

Importance  Cannabis and cannabinoid drugs are widely used to treat disease or alleviate symptoms, but their efficacy for specific indications is not clear.

Objective  To conduct a systematic review of the benefits and adverse events (AEs) of cannabinoids.

Data Sources  Twenty-eight databases from inception to April 2015.

Study Selection  Randomized clinical trials of cannabinoids for the following indications: nausea and vomiting due to chemotherapy, appetite stimulation in HIV/AIDS, chronic pain, spasticity due to multiple sclerosis or paraplegia, depression, anxiety disorder, sleep disorder, psychosis, glaucoma, or Tourette syndrome.

Data Extraction and Synthesis  Study quality was assessed using the Cochrane risk of bias tool. All review stages were conducted independently by 2 reviewers. Where possible, data were pooled using random-effects meta-analysis.

Main Outcomes and Measures  Patient-relevant/disease-specific outcomes, activities of daily living, quality of life, global impression of change, and AEs.

Results  A total of 79 trials (6462 participants) were included; 4 were judged at low risk of bias. Most trials showed improvement in symptoms associated with cannabinoids but these associations did not reach statistical significance in all trials. Compared with placebo, cannabinoids were associated with a greater average number of patients showing a complete nausea and vomiting response (47% vs 20%; odds ratio [OR], 3.82 [95% CI, 1.55-9.42]; 3 trials), reduction in pain (37% vs 31%; OR, 1.41 [95% CI, 0.99-2.00]; 8 trials), a greater average reduction in numerical rating scale pain assessment (on a 0-10-point scale; weighted mean difference [WMD], −0.46 [95% CI, −0.80 to −0.11]; 6 trials), and average reduction in the Ashworth spasticity scale (WMD, −0.12 [95% CI, −0.24 to 0.01]; 5 trials). There was an increased risk of short-term AEs with cannabinoids, including serious AEs. Common AEs included dizziness, dry mouth, nausea, fatigue, somnolence, euphoria, vomiting, disorientation, drowsiness, confusion, loss of balance, and hallucination.

Conclusions and Relevance  There was moderate-quality evidence to support the use of cannabinoids for the treatment of chronic pain and spasticity. There was low-quality evidence suggesting that cannabinoids were associated with improvements in nausea and vomiting due to chemotherapy, weight gain in HIV infection, sleep disorders, and Tourette syndrome. Cannabinoids were associated with an increased risk of short-term AEs.

Introduction

Cannabis is a generic term used for drugs produced from plants belonging to the genus Cannabis.1 It is one of the most popular recreational drugs; worldwide, an estimated 178 million people aged 15 to 64 years used cannabis at least once in 2012.2 Cannabis was included as a controlled drug in the United Nations’ Single Convention on Narcotic Drugs, held in 1961,3 and its use is illegal in most countries.

Medical cannabis refers to the use of cannabis or cannabinoids as medical therapy to treat disease or alleviate symptoms. Cannabinoids can be administered orally, sublingually,or topically; they can be smoked, inhaled, mixed with food, or made into tea. They can be taken in herbal form, extracted naturally from the plant, gained by isomerisation of cannabidiol, or manufactured synthetically.4 Prescribed cannabinoids include dronabinol capsules, nabilone capsules, and the oromucosal spray nabiximols.4 Some countries have legalized medicinal-grade cannabis for chronically ill patients. Canada and the Netherlands have government-run programs in which specialized companies supply quality-controlled herbal cannabis.5 In the United States, 23 states and Washington, DC (May 2015), have introduced laws to permit the medical use of cannabis6; other countries have similar laws. The aim of this systematic review was to evaluate the evidence for the benefits and adverse events (AEs) of medical cannabinoids across a broad range of indications.

Methods

This review followed guidance published by the Centre for Reviews and Dissemination and the Cochrane Collaboration.7,8 We established a protocol for the review (eAppendix 1 in Supplement 1).

Study Eligibility Criteria

Randomized clinical trials (RCTs) that compared cannabinoids with usual care, placebo, or no treatment in the following indications were eligible: nausea and vomiting due to chemotherapy, appetite stimulation in HIV/AIDS, chronic pain, spasticity due to multiple sclerosis (MS) or paraplegia, depression, anxiety disorder, sleep disorder, psychosis, intraocular pressure in glaucoma, or Tourette syndrome. These indications were prespecified by the project funders, the Swiss Federal Office of Public Health. If no RCTs were available for a particular indication or outcome (eg, long-term AEs such as cancer, psychosis, depression, or suicide), nonrandomized studies including uncontrolled studies (such as case series) with at least 25 patients were eligible.

Identification and Selection of Studies

Twenty-eight databases and gray literature sources were searched from inception to April 2015 without language restriction (Embase search strategy and details of databases searched available in eAppendix 2 in Supplement 2). The search strategy was peer reviewed9 by a second information specialist. Reference lists of included studies were screened. Search results and full-text articles were independently assessed by 2 reviewers; disagreements were resolved through consensus or referral to a third reviewer.

Data Collection and Study Appraisal

We extracted data about baseline characteristics and outcomes (patient-relevant and disease-specific outcomes, activities of daily living, quality of life, global impression of change, and specified AEs). For dichotomous data such as number of patients with at least 30% improvement in pain, we calculated the odds ratio (OR) and 95% CI. For categorical data, we extracted details about each category assessed and the numbers of patients with an outcome in each category. Continuous data such as the Ashworth spasticity score10 were extracted as means and SDs at baseline, follow-up, and the change from baseline and used to calculate mean differences with 95% CIs. Results (mean difference, 95% CIs, and P values) from the between-group statistical analyses reported by the study were also extracted. All relevant sources were used for data extraction including full-text journal articles, abstracts, and clinical trial registry entries. Where available, the journal article was used as the primary publication because it had been peer reviewed.

RCTs were assessed for methodological quality using the Cochrane Risk of Bias tool.11 If at least one of the domains was rated as high, the trial was considered at high risk of bias. If all domains were judged as low, the trial was considered at low risk of bias. Otherwise, the trial was considered as having unclear risk of bias. Data extraction and risk-of-bias assessment were performed independently by 2 reviewers; disagreements were resolved by a third reviewer.

Synthesis

Clinical heterogeneity was assessed by grouping studies by indication, cannabinoid, and outcome. If there were 2 or more trials within a single grouping, data were pooled using random-effects meta-analysis.12 For continuous outcomes, we analyzed the mean difference in change from baseline; if this was not reported and could not be calculated from other data, we used the mean difference at follow-up.13 For dichotomous data, we used the OR. In order to avoid double counting, we selected a single data set from each study to contribute to the analysis. For studies evaluating multiple interventions, we selected the intervention or dose that was most similar to the other interventions being evaluated in the same analysis. Heterogeneity was investigated using forest plots and the I2 statistic. Where data were considered too heterogeneous to pool or not reported in a format suitable for pooling (eg, data reported as medians), we used a narrative synthesis.

Sensitivity analyses were used to assess the statistical effect of trial design. The primary analysis included only parallel-group trials, results from crossover trials were included in an additional analysis. For the analysis of AEs, data for all conditions were combined. We conducted stratified analyses and meta-regression to investigate whether associations varied according to type of cannabinoid, study design (parallel group vs crossover trial), indication (each of the indication categories included in this report), comparator (active vs placebo), and duration of follow-up (<24 hours, 24 hours-1 week, >1 week-4 weeks, >4 weeks) for the outcome of any AE. Statistical analyses were performed using Stata statistical software (version 10).

GRADE (Grading of Recommendations Assessment, Development and Evaluation) was used to rate the overall quality of the evidence for risk of bias, publication bias, imprecision, inconsistency, indirectness, and magnitude of effect. The GRADE ratings of very low–, low-, moderate-, or high-quality evidence reflect the extent to which we are confident that the effect estimates are correct.14

Results

The searches identified 23 754 hits (records) of which 505 were considered potentially relevant, based on title and abstract screening, and obtained as full-text studies. A total of 79 studies (6462 participants), available as 151 reports, were included; 3 studies (6 reports) were included in multiple indication categories (Figure 1). Thirty-four studies were parallel-group trials (4436 participants), and 45 were crossover trials (2026 participants). Four studies were available only as an abstract,1518 a further 3 were available only as abstracts1921 but with additional details available on trial registries including full results in one,19 and details of 2 trials (including full trial results) were available only as trial registry entries22,23; all other trials were reported in full-length journal articles. Where reported, the proportion of participants who were men ranged from 0% to 100% (median, 50% [57 studies]), and the proportion of white participants ranged from 50% to 99% (median, 78% [18 studies]). Publication dates ranged from 1975 to 2015 (median, 2004 [with one-third of trials published before 1990]). Studies were conducted in a wide range of countries. A variety of cannabinoids were evaluated and compared with various different active comparators or placebos; most active comparators were included in the nausea and vomiting indication (Table 1). eAppendices 3 to 12 in Supplement 1 provide an overview of the included studies and their findings.

Four (5%) trials were judged at low risk of bias, 55 (70%) were judged at high risk of bias, and 20 (25%) at unclear risk of bias (eAppendix 13 in Supplement 2). The major potential source of bias in the trials was incomplete outcome data. More than 50% of trials reported substantial withdrawals and did not adequately account for this in the analysis. Selective outcome reporting was a potential risk of bias in 16% of trials. These studies did not report data for all outcomes specified in the trial register, protocol, or methods section or changed the primary outcome from that which was prespecified. Most studies reported being double-blinded but only 57% reported that appropriate methods had been used for participant blinding and only 24% reported that outcome assessors had been appropriately blinded.

Full results from included studies are presented in eAppendices 3-12 in Supplement 2; pooled results and GRADE ratings are presented in Table 2.

Nausea and Vomiting Due to Chemotherapy

Nausea and vomiting due to chemotherapy was assessed in 28 studies (37 reports; 1772 participants).15,16,2458 Fourteen studies assessed nabilone and there were 3 for dronabinol, 1 for nabiximols, 4 for levonantradol, and 6 for THC. Two studies also included a combination therapy group of dronabinol with ondansetron or prochlorperazine. Eight studies included a placebo control, 3 of these also included an active comparator, and 20 studies included only an active comparator. The most common active comparators were prochlorperazine (15 studies), chlorpromazine (2 studies) and domperidone (2 studies). Other comparators (alizapride, hydroxyzine, metoclopramide and ondansetron) were evaluated in single studies (Table 1). Of all 28 studies, risk of bias was high for 23 or unclear for 5. All studies suggested a greater benefit of cannabinoids compared with both active comparators and placebo, but these did not reach statistical significance in all studies. The average number of patients showing a complete nausea and vomiting response was greater with cannabinoids (dronabinol or nabiximols) than placebo (OR, 3.82 [95% CI, 1.55-9.42]; 3 trials). There was no evidence of heterogeneity for this analysis (I2 = 0%) and results were similar for both dronabinol and nabiximols.

Appetite Stimulation in HIV/AIDS Infection

Appetite stimulation in HIV/AIDS was assessed in 4 studies (4 reports; 255 participants).5962 All studies assessed dronabinol, 3 compared with placebo (1 of which also assessed marijuana), and 1 compared with megastrol acetate. All studies were at high risk of bias. There was some evidence that dronabinol is associated with an increase in weight when compared with placebo. More limited evidence suggested that it may also be associated with increased appetite, greater percentage of body fat, reduced nausea, and improved functional status. However, these outcomes were mostly assessed in single studies and associations failed to reach statistical significance. The trial that evaluated marijuana and dronabinol found significantly greater weight gain with both forms of cannabinoid when compared with placebo.59 The active comparison trial found that megastrol acetate was associated with greater weight gain than dronabinol and that combining dronabinol with megastrol acetate did not lead to additional weight gain.60

Chronic Pain

Chronic pain was assessed in 28 studies (63 reports; 2454 participants).19,20,22,23,63120 Thirteen studies evaluated nabiximols, 4 were for smoked THC, 5 for nabilone, 3 for THC oromucosal spray, 2 dronabinol, 1 vaporized cannabis (included 2 doses), 1 for ajuvenic acid capsules, and 1 for oral THC. One trial compared nabilone with amitriptyline64; all other studies were placebo controlled. One of these studies evaluated nabilone as an adjunctive treatment to gabapentin.121 The conditions causing the chronic pain varied between studies and included neuropathic pain (central, peripheral, or not specified; 12 studies), 3 for cancer pain, 3 for diabetic peripheral neuropathy, 2 for fibromyalgia, 2 for HIV-associated sensory neuropathy, and 1 study for each of the following indications: refractory pain due to MS or other neurological conditions, for rheumatoid arthritis, for noncancer pain (nociceptive and neuropathic), central pain (not specified further), musculoskeletal problems, and chemotherapy-induced pain.

Two studies were at low risk of bias, 9 at unclear risk, and 17 at high risk of bias. Studies generally suggested improvements in pain measures associated with cannabinoids but these did not reach statistical significance in most individual studies.

The average number of patients who reported a reduction in pain of at least 30% was greater with cannabinoids than with placebo (OR, 1.41 [95% CI, 0.99-2.00]; 8 trials; Figure 2). One trial assessed smoked THC77 and reported the greatest beneficial effect (OR, 3.43 [95% CI, 1.03-11.48]), and 7 trials assessed nabiximols (Figure 2). Pain conditions evaluated in these trials were neuropathic pain (OR, 1.38 [95% CI, 0.93-2.03]; 6 trials) and cancer pain (OR, 1.41 [95% CI, 0.99-2.00]; 2 trials), with no clear differences between pain conditions. Nabiximols was also associated with a greater average reduction in the Numerical Rating Scale (NRS; 0-10 scale) assessment of pain (weighted mean difference [WMD], −0.46 [95% CI, −0.80 to −0.11]; 6 trials), brief pain inventory-short form, severity composite index (WMD, −0.17 [95% CI, −0.50 to 0.16]; 3 trials), neuropathic pain scale (WMD, −3.89 [95% CI, −7.32 to −0.47]; 5 trials), and the proportion of patients reporting improvement on a global impression of change score (OR, 2.08 [95% CI, 1.21 to 3.59]; 6 trials) compared with placebo. There was some evidence to support this based on continuous data but this was not consistent across trials. There was no difference in average quality-of-life scores as measured by the EQ-5D health status index (WMD, −0.01 [95% CI, −0.05 to 0.02]; 3 trials) between nabiximols and placebo. Two of the studies included in the meta-analysis for the NRS (0-10 scale) assessed patients with cancer pain, all other studies assessed patients with neuropathic pain. There were no clear differences based on cause of pain in the meta-analysis of NRS. Sensitivity analyses that included crossover trials showed results consistent with those based on parallel-group trials alone.

Spasticity Due to MS or Paraplegia

Fourteen studies (33 reports; 2280 participants) assessed spasticity due to MS or paraplegia.17,19,65,87,91,122149 Eleven studies (2138 participants) included patients with MS and 3 included patients with paraplegia (142 participants) caused by spinal cord injury. Six studies assessed nabiximols, 3 for dronabinol, 1 for nabilone, 4 for THC/CBD (2 of these also assessed dronabinol), and 1 each for ECP002A and smoked THC. All studies included a placebo control group; none included an active comparator. Two studies were at low risk of bias, 5 were at unclear risk of bias, and 7 were at high risk of bias. Studies generally suggested that cannabinoids were associated with improvements in spasticity, but this failed to reach statistical significance in most studies. There were no clear differences based on type of cannabinoid. Only studies in MS patients reported sufficient data to allow summary estimates to be generated. Cannabinoids (nabiximols, dronabinol, and THC/CBD) were associated with a greater average improvement on the Ashworth scale for spasticity compared with placebo, although this did not reach statistical significance (WMD, −0.12 [95% CI, −0.24 to 0.01]; 5 trials; Figure 3). Cannabinoids (nabilone and nabiximols) were also associated with a greater average improvement in spasticity assessed using numerical rating scales (mean difference, −0.76 [95% CI, −1.38 to −0.14]; 3 trials). There was no evidence of a difference in association according to type of cannabinoid for either analysis. Other measures of spasticity also suggested a greater benefit of cannabinoid but did not reach statistical significance (Table 2). The average number of patients who reported an improvement on a global impression of change score was also greater with nabiximols than placebo (OR, 1.44 [95% CI, 1.07 to 1.94]; 3 trials); this was supported by a further crossover trial of dronabinol and oral THC/CBD that provided continuous data for this outcome.132 Sensitivity analyses that included crossover trials showed results consistent with those based on parallel group trials alone.

Depression

No studies evaluating cannabinoids for the treatment of depression fulfilled inclusion criteria. Five studies included for other indications reported depression as an outcome measure; 4 evaluated chronic pain and 1 evaluated spasticity in MS patients.67,73,75,80,129 One trial assessed dronabinol (2 doses), 3 assessed nabiximols, and 1 assessed nabilone. Two studies were rated as having unclear risk of bias and 3 as having high risk of bias. Three studies suggested no difference between cannabinoids (dronabinol and nabiximols) and placebo in depression outcomes. One parallel-group trial that compared different doses of nabiximols with placebo reported a negative effect of nabiximols for the highest dose (11-14 sprays per day) compared with placebo (mean difference from baseline, 2.50 [95% CI, 0.38 to 4.62]) but no difference between placebo and the 2 lower doses.67

Anxiety Disorder

One small parallel-group trial, judged at high risk of bias, evaluated patients with generalized social anxiety disorder.150 The trial reported that cannabidiol was associated with a greater improvement on the anxiety factor of a visual analogue mood scale (mean difference from baseline, −16.52; P value = .01)compared with placebo during a simulated public speaking test. Additional data about anxiety outcomes provided by 4 studies (1 parallel group) in patients with chronic pain also suggested a greater benefit of cannabinoids (dronabinol, nabilone, and nabiximols) than placebo but these studies were not restricted to patients with anxiety disorders.7375,80

Sleep Disorder

Two studies (5 reports; 54 participants) evaluated cannabinoids (nabilone) specifically for the treatment of sleep problems. One was a parallel-group trial judged at high risk of bias. This reported a a greater benefit of nabilone compared with placebo on the sleep apnea/hypopnea index (mean difference from baseline, −19.64; P value = .02). The other was a crossover trial judged at low risk of bias in patients with fibromyalgia and compared nabilone with amitriptyline. This suggested that nabilone was associated with improvements in insomnia (mean difference from baseline, −3.25 [95% CI, −5.26 to −1.24]) and with greater sleep restfulness (mean difference from baseline, 0.48 [95% CI, 0.01 to 0.95]). Nineteen placebo-controlled studies included for other indications (chronic pain and MS) also evaluated sleep as an outcome.22,23,65,6769,75,76,7981,87,88,123125,129131 Thirteen studies assessed nabiximols, 1 for nabilone, 1 for dronabinol, 2 for THC/CBD capsules, and two assessed smoked THC (one at various doses). Two of the studies that assessed nabiximols also assessed oral THC and the trial of dronabinol also assessed oral THC/CBD. There was some evidence that cannabinoids may improve sleep in these patient groups. Cannabinoids (mainly nabiximols) were associated with a greater average improvement in sleep quality (WMD, −0.58 [95% CI, −0.87 to −0.29]; 8 trials) and sleep disturbance (WMD, −0.26 [95% CI, −0.52 to 0.00]; 3 trials). One trial assessed THC/CBD, all others assessed nabiximols, results were similar for both cannabinoids.

Psychosis

Psychosis was assessed in 2 studies (9 reports; 71 participants) judged at high risk of bias, which evaluated cannabidiol compared with amisulpride or placebo.21,151158 The trials found no difference in mental health outcomes between treatment groups.

Glaucoma

One very small crossover trial (6 participants)159 judged at unclear risk of bias compared tetrahydrocannabinol (THC; 5 mg), cannabidiol (20 mg), cannabidiol (40 mg) oromucosal spray, and placebo. This trial found no difference between placebo and cannabinoids on measures of intraocular pressure in patients with glaucoma.

Movement Disorders Due to Tourette Syndrome

Two small placebo-controlled studies (4 reports; 36 participants)160163 suggested that THC capsules may be associated with a significant improvement in tic severity in patients with Tourette syndrome.

Adverse Events

Data about AEs were reported in 62 studies (127 reports). Meta-regression and stratified analysis showed no evidence for a difference in the association of cannabinoids with the incidence of “any AE” based on type of cannabinoid, study design, indication, comparator, or duration of follow-up15,16,18,2226,2831,3338,41,42,4447,51,57,58,60,62,6469,7285,87,88,123127,129131,159,160,162; further analyses were conducted for all studies combined. Figure 4 shows the results of the meta-analyses for the number of participants experiencing any AE compared when compared with controls, stratified according to cannabinoid. Cannabinoids were associated with a much greater risk of any AE, serious AE, withdrawals due to AE, and a number of specific AEs (Table 3). No studies evaluating the long-term AEs of cannabinoids were identified, even when searches were extended to lower levels of evidence.

Discussion

We conducted an extensive systematic review of the benefits and AEs associated with medical cannabinoids across a broad range of conditions. We included 79 RCTs (6462 participants), the majority of which evaluated nausea and vomiting due to chemotherapy or chronic pain and spasticity due to MS and paraplegia. Other patient categories were evaluated in fewer than 5 studies.

Most studies suggested that cannabinoids were associated with improvements in symptoms, but these associations did not reach statistical significance in all studies. Based on the GRADE approach, there was moderate-quality evidence to suggest that cannabinoids may be beneficial for the treatment of chronic neuropathic or cancer pain (smoked THC and nabiximols) and spasticity due to MS (nabiximols, nabilone, THC/CBD capsules, and dronabinol). There was low-quality evidence suggesting that cannabinoids were associated with improvements in nausea and vomiting due to chemotherapy (dronabinol and nabiximols), weight gain in HIV (dronabinol), sleep disorders (nabilone, nabiximols), and Tourette syndrome (THC capsules); and very low-quality evidence for an improvement in anxiety as assessed by a public speaking test (cannabidiol). There was low-quality evidence for no effect on psychosis (cannabidiol) and very low-level evidence for no effect on depression (nabiximols). There was an increased risk of short-term AEs with cannabinoid use, including serious AEs. Common AEs included asthenia, balance problems, confusion, dizziness, disorientation, diarrhea, euphoria, drowsiness, dry mouth, fatigue, hallucination, nausea, somnolence, and vomiting. There was no clear evidence for a difference in association (either beneficial or harmful) based on type of cannabinoids or mode of administration. Only 2 studies evaluated cannabis.59,77 There was no evidence that the effects of cannabis differed from other cannabinoids.

Strengths and Weaknesses

This review followed recommendations for rigorous systematic reviews.7,8 In order to identify as many relevant studies as possible and reduce the risk of publication bias, a highly sensitive search strategy was used and an extensive range of resources were searched including electronic databases, guidelines, and systematic reviews. Both published and unpublished trials were eligible for inclusion. There were no date or language restrictions. In order to minimize bias and errors, the main Embase strategies were peer reviewed by a second independent information specialist165 and all stages of the review process were performed independently by 2 reviewers. We used the Cochrane risk of bias tool11 to assess the included RCTs. This highlighted a number of methodological weaknesses in the included trials including failure to appropriately handle withdrawals, selective outcome reporting, and inadequate description of methods of randomization, allocation concealment, and blinding. An additional limitation of many included studies was their very small sample sizes. This was particularly the case for the trial of glaucoma (N = 6), Tourette syndrome (average N = 18), sleep disorder (average N = 27), and anxiety disorder (N = 24), which means these studies may have lacked the power to detect differences between treatment groups.

The synthesis combined a narrative discussion of individual study results with meta-analysis (for studies in which suitable data were available), supplemented by interpretation (following guidance of the GRADE Working Group).14 The data analysis was complicated by a number of issues. The included studies used a large variety of measures to evaluate outcomes, and even very similar outcomes were often assessed using different measures. Furthermore, a wide range of time points were reported in the included trials, which limited the applicability of the findings of these studies. Multiple different cannabinoids were evaluated in the included studies. We stratified analyses based on type of cannabinoid to investigate whether there were differences in associations based on type of cannabinoid. The majority of the studies were 2-group trials with a placebo control group; however, some studies included active comparisons and multiple groups comparing more than 1 form of cannabinoid, different doses of cannabinoids, or active and placebo comparator groups. This necessitated selecting a single result from each trial to contribute to the meta-analysis to avoid double counting of studies. Where possible, we selected the result for the treatment or dose most similar to the other studies contributing to that meta-analysis and for placebo-controlled comparisons rather than active comparisons. For the short-term AE analysis, we selected the highest-reported cannabinoids dose because we hypothesized that this would be most likely to be associated with AEs—additionally, this analysis would present a worst-case scenario. Studies evaluated various forms of cannabis administered via various routes (oral capsules, smoked, vaporized, oromucosal spray, intramuscular injection) and active comparators differed across trials. These differences in form, combined with the variety of outcome measures and the broad indication groupings considered by this review, resulted in a very heterogeneous set of included studies, which meant that meta-analysis was not always possible or appropriate. Many studies reported insufficient information to allow meta-analysis (eg, reporting only P values for group differences) or no information on the analysis performed. A further difficulty with the continuous data were that even for the same outcomes, some studies reported results as difference between groups at follow-up and others reported results for difference in change from baseline. As advised by the Cochrane Handbook for Systematic Reviews of Interventions, we combined both types of data when estimating summary mean differences.7 A potential problem with RCTs using crossover designs is the possible unblinding due to strong treatment or AEs. Additionally, studies of this design were rarely analyzed appropriately and none reported the required data accounting for their crossover design to permit appropriate inclusion in meta-analyses.166 Primary analyses were therefore based on parallel-group studies, with crossover trials included as sensitivity analyses.

Our search identified a number of existing reviews that assessed the use of medical cannabinoids for MS,167170 nausea and vomiting due to chemotherapy,171175 pain,176191 psychosis,192194 and Tourette syndrome.195,196 Almost all previous reviews focused on single indications and all but one (which evaluated cannabinoids in 4 trials in patients with pain due to rheumatoid arthritis)188 did not use the GRADE approach to rating the quality of the evidence. As far as we are aware, our review is the first comprehensive review to evaluate the safety and efficacy of cannabinoids across a broad range of indications. A key strength of review was that it allowed us to conduct pooled analysis for the AEs associated with medicinal cannabinoids, adding considerable power to this analysis.

Unanswered Questions and Future Research

Further large, robust, RCTs are needed to confirm the effects of cannabinoids, particularly on weight gain in patients with HIV/AIDS, depression, sleep disorders, anxiety disorders, psychosis, glaucoma, and Tourette syndrome are required. Further studies evaluating cannabis itself are also required because there is very little evidence on the effects and AEs of cannabis. Future trials should adhere to the CONSORT (Consolidated Standards of Reporting Trials) reporting standards197 and ensure that appropriate methods are used for randomization, allocation concealment, patient and outcome assessor blinding, handling of withdrawals, and avoiding selective outcome reporting. Future studies should assess patient-relevant outcomes (including disease-specific end points, quality of life, and AEs) using standardized outcome measures at similar time points to ensure inclusion in future meta-analyses.

Conclusions

There was moderate-quality evidence to support the use of cannabinoids for the treatment of chronic pain and spasticity. There was low-quality evidence suggesting that cannabinoids were associated with improvements in nausea and vomiting due to chemotherapy, weight gain in HIV, sleep disorders, and Tourette syndrome. Cannabinoids were associated with an increased risk of short-term AEs.

Back to top
Article Information

Corresponding Author: Penny Whiting, PhD, NIHR CLAHRC West, University Hospitals Bristol NHS Foundation Trust, Ninth Floor, Whitefriars, Lewins Mead, Bristol BS1 2NT, United Kingdom (penny.whiting@bristol.ac.uk).

Author Contributions: Dr Whiting had full access to all of the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis.

Study concept and design: Whiting, Wolff, Misso, Kleijnen.

Acquisition, analysis, or interpretation of data: Whiting, Wolff, Deshpande, Di Nisio, Duffy, Hernandez, Keurentjes, Lang, Misso, Ryder, Schmidlkofer, Westwood.

Drafting of the manuscript: Whiting, Keurentjes, Ryder.

Critical revision of the manuscript for important intellectual content: Whiting, Wolff, Deshpande, Di Nisio, Duffy, Hernandez, Keurentjes, Lang, Misso, Ryder, Schmidlkofer, Westwood, Kleijnen.

Statistical analysis: Whiting, Wolff, Di Nisio, Hernandez, Keurentjes, Schmidlkofer.

Obtained funding: Kleijnen.

Administrative, technical, or material support: Deshpande, Lang, Ryder.

Study supervision: Whiting, Kleijnen.

Conflict of Interest Disclosures: All authors have completed and submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest and declare support from the Swiss Federal Office of Public Health (FOPH) for the submitted work; no financial relationships with any organizations that might have an interest in the submitted work in the previous 3 years; and no other relationships or activities that could appear to have influenced the submitted work. Dr Whiting reports that part of her time on this review was supported by the National Institute for Health Research (NIHR) Collaboration for Leadership in Applied Health Research and Care West at University Hospitals Bristol NHS (National Health Service) Foundation Trust. No additional disclosures were reported.

Funding/Support: This funded by the Swiss Federal Office of Public Health (FOPH) under grant agreement 14.001443/204.0001/-1257.

Role of the Funder/Sponsor: The FOPH had no role in the design and conduct of the study; collection, management, analysis, and interpretation of the data; preparation, review, or approval of the manuscript; and decision to submit the manuscript for publication. The decision to submit the article for publication was a condition of the funding and was made before any results were available.

Additional Author Contributions: Dr Whiting drafted the article, produced tables and figures and performed the analysis. Drs Whiting, Wolff, and Kleijnen and Ms Misso and Mr Duffy drafted the protocol. Mr Duffy and Ms Misso conducted the literature searches. Drs Whiting, Wolff, and Lang screened searched results and selected full-text studies for inclusion. Drs Whiting, Wolff, Lang, Westwood, Keurentjes, Di Nisio, Hernandez, and Messrs Deshpande and Ryder, and Ms Schmidlkofer performed data extraction and risk-of-bias assessment. Dr Wolff performed the GRADE assessments. All authors agree to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Disclaimer: The views expressed are those of the author and not necessarily those of the NHS, the NIHR or the Department of Health.

Additional Contributions: We would like to thank Julie Harker (MRes, Kleijnen Systematic Reviews at the time of this project) for help with inclusion screening and data extraction and Gillian Worthy (MSc, Kleijnen Systematic Reviews) for advice on data analysis. Neither of these individuals received additional compensation in association with their work on this article.

Correction: This article was corrected online June 26, 2015, for incorrect axis labeling in Figure 4; on July 13, 2015, for a corrected average reduction to the Ashworth spasticity scale (as reported in the Abstract); on November 5, 2015, for an incorrect nonproprietary name and approved use for a drug in Table 1; and on April 12, 2016, for an incorrect effect estimate.

References
1.
Small  E, Cronquist  A.  A practical and natural taxonomy for cannabis.  Taxon. 1976;25(4):405-435. doi:10.2307/1220524.Google ScholarCrossref
2.
Poznyak  V.  SY14-1 global epidemiology of cannabis use and implications for public health.  Alcohol Alcohol. 2014;49(suppl 1):i14. doi:10.1093/alcalc/agu052.58 i.Google ScholarCrossref
3.
United Nations.  Single Convention on Narcotic Drugs, 1961. New York, NY: United Nations; 1962.
4.
Hazekamp  A, Ware  MA, Müller-Vahl  KR, Abrams  D, Grotenhermen  F.  The medicinal use of cannabis and cannabinoids—an international cross-sectional survey on administration forms.  J Psychoactive Drugs. 2013;45(3):199-210.PubMedGoogle ScholarCrossref
5.
Hazekamp  A, Heerdink  ER.  The prevalence and incidence of medicinal cannabis on prescription in the Netherlands.  Eur J Clin Pharmacol. 2013;69(8):1575-1580.PubMedGoogle ScholarCrossref
6.
Office of National Drug Control Policy.  Marijuana Resource Center: State Laws Related to Marijuana.https://www.whitehouse.gov/ondcp/state-laws-related-to-marijuana. Accessed May 18, 2015.
7.
Higgins  JPT, Green  S, eds.  Cochrane Handbook for Systematic Reviews of Interventions. Version 5.1.0 (updated March 2011). The Cochrane Collaboration website. http://handbook.cochrane.org/. Accessed March 23, 2011.
8.
Centre for Reviews and Dissemination.  Systematic Reviews: CRD’s Guidance for Undertaking Reviews in Health Care (Internet). York, England: University of York; 2009. https://www.york.ac.uk/media/crd/Systematic_Reviews.pdf. Accessed March 23, 2011.
9.
Canadian Agency for Drugs and Technologies in Health.  CADTH Peer Review Checklist for Search Strategies (Internet). Ottawa, Canada: CADTH; 2013. https://www.cadth.ca/resources/finding-evidence/cadth-peer-review-checklist-search-strategies. Accessed March 17, 2014.
10.
Bohannon  RW, Smith  MB.  Interrater reliability of a modified Ashworth scale of muscle spasticity.  Phys Ther. 1987;67(2):206-207.PubMedGoogle Scholar
11.
Higgins  JP, Altman  DG, Gøtzsche  PC,  et al; Cochrane Bias Methods Group; Cochrane Statistical Methods Group.  The Cochrane Collaboration’s tool for assessing risk of bias in randomised trials.  BMJ. 2011;343:d5928.PubMedGoogle ScholarCrossref
12.
DerSimonian  R, Laird  N.  Meta-analysis in clinical trials.  Control Clin Trials. 1986;7(3):177-188.PubMedGoogle ScholarCrossref
13.
Higgins  JP, Thompson  SG.  Quantifying heterogeneity in a meta-analysis.  Stat Med. 2002;21(11):1539-1558.PubMedGoogle ScholarCrossref
14.
Guyatt  GH, Oxman  AD, Vist  G,  et al.  GRADE guidelines: 4. rating the quality of evidence—study limitations (risk of bias).  J Clin Epidemiol. 2011;64(4):407-415.PubMedGoogle ScholarCrossref
15.
Broder  LE, Lean  NL, Hilsenbeck  SG.  A randomized blinded clinical trial comparing delta-9-tetrahydrocannabinol (THC) and hydroxizine (HZ) as antiemetics (AE) for cancer chemotherapy (CT).  Proc Am Assoc Cancer Res. 1982;23:514.Google Scholar
16.
Long  A, Mioduszewski  J, Natale  R.  A randomized double-blind cross-over comparison of the antiemetic activity of levonantradol and prochlorperazine.  Proc Am Soc Clin Oncol. 1982;1:C-220.Google Scholar
17.
Hagenbach  U, Luz  S, Brenneisen  R, Mäder  M.  The treatment of spasticity with D9-tetrahydrocannabinol (D9-THC) in patients with spinal cord injury. Paper presented at: IACM 2nd Conference on Cannabinoids in Medicine; September 12-13, 2003; Cologne, Germany.
18.
Prasad  B, Radulovacki  MG, Carley  DW.  Randomized placebo controlled trial of dronabinol in obstructive sleep apnea. Paper presented at: American Thoracic Society International Conference, ATS 2011; May 13-18, 2011; Denver, CO. Am J Respir Crit Care Med. 2011;183(1):A2720.
19.
GW Pharmaceuticals Ltd.  A study of cannabis based medicine extracts and placebo in patients with pain due to spinal cord injury. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT01606202. Accessed April 7, 2014.
20.
Center for Medicinal Cannabis Research.  Efficacy of inhaled cannabis in diabetic painful peripheral neuropathy. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00781001. Accessed April 7, 2014.
21.
Stanley Medical Research Institute, Coordinating Centre for Clinical Trials Cologne.  University of Cologne. A clinical trial on the antipsychotic properties of cannabidiol. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00309413. Accessed April 7, 2014.
22.
GW Pharma Ltd.  A double blind, randomised, placebo controlled, parallel group study of Sativex in the treatment of subjects with pain due to diabetic neuropathy. EU Clinical Trials Register. https://www.clinicaltrialsregister.eu/ctr-search/search?query=eudract_number:2004-002530-20. Accessed August 4, 2014.
23.
GW Pharmaceuticals Ltd.  A study to evaluate the effects of cannabis based medicine in patients with pain of neurological origin. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT01606176 Accessed April 7, 2014.
24.
Duran  M, Pérez  E, Abanades  S,  et al.  Preliminary efficacy and safety of an oromucosal standardized cannabis extract in chemotherapy-induced nausea and vomiting.  Br J Clin Pharmacol. 2010;70(5):656-663.PubMedGoogle ScholarCrossref
25.
Meiri  E, Jhangiani  H, Vredenburgh  JJ,  et al.  Efficacy of dronabinol alone and in combination with ondansetron versus ondansetron alone for delayed chemotherapy-induced nausea and vomiting.  Curr Med Res Opin. 2007;23(3):533-543.PubMedGoogle ScholarCrossref
26.
Lane  M, Vogel  CL, Ferguson  J,  et al.  Dronabinol and prochlorperazine in combination for treatment of cancer chemotherapy-induced nausea and vomiting.  J Pain Symptom Manage. 1991;6(6):352-359.PubMedGoogle ScholarCrossref
27.
McCabe  M, Smith  FP, Macdonald  JS, Woolley  PV, Goldberg  D, Schein  PS.  Efficacy of tetrahydrocannabinol in patients refractory to standard antiemetic therapy.  Invest New Drugs. 1988;6(3):243-246.PubMedGoogle ScholarCrossref
28.
Chan  HS, Correia  JA, MacLeod  SM.  Nabilone versus prochlorperazine for control of cancer chemotherapy-induced emesis in children: a double-blind, crossover trial.  Pediatrics. 1987;79(6):946-952.PubMedGoogle Scholar
29.
Pomeroy  M, Fennelly  JJ, Towers  M.  Prospective randomized double-blind trial of nabilone versus domperidone in the treatment of cytotoxic-induced emesis.  Cancer Chemother Pharmacol. 1986;17(3):285-288.PubMedGoogle ScholarCrossref
30.
Dalzell  AM, Bartlett  H, Lilleyman  JS.  Nabilone: an alternative antiemetic for cancer chemotherapy.  Arch Dis Child. 1986;61(5):502-505.PubMedGoogle ScholarCrossref
31.
Niederle  N, Schütte  J, Schmidt  CG.  Crossover comparison of the antiemetic efficacy of nabilone and alizapride in patients with nonseminomatous testicular cancer receiving cisplatin therapy.  Klin Wochenschr. 1986;64(8):362-365.PubMedGoogle ScholarCrossref
32.
Niiranen  A, Mattson  K.  A cross-over comparison of nabilone and prochlorperazine for emesis induced by cancer chemotherapy.  Am J Clin Oncol. 1985;8(4):336-340.PubMedGoogle ScholarCrossref
33.
Heim  ME, Queisser  W, Altenburg  HP.  Randomized crossover study of the antiemetic activity of levonantradol and metoclopramide in cancer patients receiving chemotherapy.  Cancer Chemother Pharmacol. 1984;13(2):123-125.PubMedGoogle ScholarCrossref
34.
Hutcheon  AW, Palmer  JB, Soukop  M,  et al.  A randomised multicentre single blind comparison of a cannabinoid anti-emetic (levonantradol) with chlorpromazine in patients receiving their first cytotoxic chemotherapy.  Eur J Cancer Clin Oncol. 1983;19(8):1087-1090.PubMedGoogle ScholarCrossref
35.
George  M, Pejovic  MH, Thuaire  M, Kramar  A, Wolff  JP.  [Randomized comparative trial of a new anti-emetic: nabilone, in cancer patients treated with cisplatin].  Biomed Pharmacother. 1983;37(1):24-27.PubMedGoogle Scholar
36.
Jones  SE, Durant  JR, Greco  FA, Robertone  A.  A multi-institutional phase III study of nabilone vs placebo in chemotherapy-induced nausea and vomiting.  Cancer Treat Rev.1982;9(suppl B):45-48.PubMedGoogle ScholarCrossref
37.
Wada  JK, Bogdon  DL, Gunnell  JC, Hum  GJ, Gota  CH, Rieth  TE.  Double-blind, randomized, crossover trial of nabilone vs placebo in cancer chemotherapy.  Cancer Treat Rev.1982;9(suppl B):39-44.PubMedGoogle ScholarCrossref
38.
Johansson  R, Kilkku  P, Groenroos  M. A double-blind, controlled trial of nabilone vs. prochlorperazine for refractory emesis induced by cancer chemotherapy.  Cancer Treat Rev.1982;9(suppl B):25-33.PubMedGoogle ScholarCrossref
39.
Orr  LE, McKernan  JF.  Antiemetic effect of delta 9-tetrahydrocannabinol in chemotherapy-associated nausea and emesis as compared to placebo and compazine.  J Clin Pharmacol.1981;21(8-9 suppl):76S-80S.PubMedGoogle ScholarCrossref
40.
Einhorn  LH, Nagy  C, Furnas  B, Williams  SD.  Nabilone: an effective antiemetic in patients receiving cancer chemotherapy.  J Clin Pharmacol.1981;21(8-9 suppl):64S-69S.PubMedGoogle ScholarCrossref
41.
Orr  LE, McKernan  JF, Bloome  B.  Antiemetic effect of tetrahydrocannabinol: compared with placebo and prochlorperazine in chemotherapy-associated nausea and emesis.  Arch Intern Med. 1980;140(11):1431-1433.PubMedGoogle ScholarCrossref
42.
Steele  N, Gralla  RJ, Braun  DW  Jr, Young  CW.  Double-blind comparison of the antiemetic effects of nabilone and prochlorperazine on chemotherapy-induced emesis.  Cancer Treat Rep. 1980;64(2-3):219-224.PubMedGoogle Scholar
43.
Sallan  SE, Cronin  C, Zelen  M, Zinberg  NE.  Antiemetics in patients receiving chemotherapy for cancer: a randomized comparison of delta-9-tetrahydrocannabinol and prochlorperazine.  N Engl J Med. 1980;302(3):135-138.PubMedGoogle ScholarCrossref
44.
Frytak  S, Moertel  CG, O’Fallon  JR,  et al.  Delta-9-tetrahydrocannabinol as an antiemetic for patients receiving cancer chemotherapy: a comparison with prochlorperazine and a placebo.  Ann Intern Med. 1979;91(6):825-830.PubMedGoogle ScholarCrossref
45.
Ahmedzai  S, Carlyle  DL, Calder  IT, Moran  F.  Anti-emetic efficacy and toxicity of nabilone, a synthetic cannabinoid, in lung cancer chemotherapy.  Br J Cancer. 1983;48(5):657-663.PubMedGoogle ScholarCrossref
46.
Ungerleider  JT, Andrysiak  T, Fairbanks  L, Goodnight  J, Sarna  G, Jamison  K.  Cannabis and cancer chemotherapy: a comparison of oral delta-9-THC and prochlorperazine.  Cancer. 1982;50(4):636-645.PubMedGoogle ScholarCrossref
47.
Sheidler  VR, Ettinger  DS, Diasio  RB, Enterline  JP, Brown  MD.  Double-blind multiple-dose crossover study of the antiemetic effect of intramuscular levonantradol compared to prochlorperazine.  J Clin Pharmacol. 1984;24(4):155-159.PubMedGoogle ScholarCrossref
48.
Harden-Harrison  MM, Munsell  MF, Fisch  MJ,  et al.  Dronabinol for the prevention of nausea from cyclophosphamide and/or adriamycin. Paper presented at: International MASCC/ISOO Symposium: Supportive Care in Cancer; June 28-30, 2012; New York, NY. Support Care Cancer. 2012;20:S209-S210.
49.
Grunberg  SM, Munsell  MF, Morrow  PKH,  et al.  Randomized double-blind evaluation of dronabinol for the prevention of chemotherapy-induced nausea. Paper presented at: Annual Meeting of the American Society of Clinical Oncology (ASCO); 1-5 Jun 2012; Chicago, IL. J Clin Oncol. 2012;30(15)(suppl 1):9061.
50.
Lane  M, Vogel  CL, Ferguson  J,  et al.  Dronabinol and prochlorperazine in combination are better than either single agent alone for treatment of chemotherapy-induced nausea and vomiting.  Proc Am Soc Clin Oncol. 1989;8:326.Google Scholar
51.
Levitt  M. Nabilone vs placebo in the treatment of chemotherapy-induced nausea and vomiting in cancer patients.  Cancer Treat Rev.1982;9(suppl B):49-53.PubMedGoogle ScholarCrossref
52.
Chan  HS, MacLeod  SM, Correia  JA.  Nabilone vs prochlorperazine for control of cancer chemotherapy-induced emesis in children.  Proc Am Soc Clin Oncol. 1984;3:108.Google Scholar
53.
Solvay Pharmaceuticals.  Dronabinol versus standard ondansetron antiemetic therapy in preventing delayed-onset chemotherapy-induced nausea and vomiting. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00642512 Accessed April 7, 2014.
54.
Frytak  S, Moertel  CG, Ofallon  JR.  Comparison of delta-9-tetrahydrocannabinol (THC), prochlorperazine (PCP) and placebo as anti-emetics for cancer-chemotherapy.  Proc Am Assoc Cancer Res. 1979;20:391.Google Scholar
55.
Jhangiani  H, Vredenburgh  JJ, Barbato  L,  et al.  Dronabinol or ondansetron alone and combined for delayed chemotherapy-induced nausea and vomiting (CINV).  Blood. 2005;106(11, part 2):477B.PubMedGoogle Scholar
56.
McCabe  M, Smith  FP, Goldberg  D,  et al.  Comparative trial of oral 9 tetrahydrocannabinol and prochlorperazine for cancer chemotherapy related nausea and vomiting.  Proc Am Assoc Cancer Res and Am Soc Clin Oncol.1981;22:416.Google Scholar
57.
Herman  TS, Einhorn  LH, Jones  SE,  et al.  Superiority of nabilone over prochlorperazine as an antiemetic in patients receiving cancer chemotherapy.  N Engl J Med. 1979;300(23):1295-1297.PubMedGoogle ScholarCrossref
58.
Melhem-Bertrandt  AI, Munsell  MF, Fisch  MJ,  et al A randomized, double-blind, placebo-controlled trial of palonosetron plus dexamethasone with or without dronabinol for the prevention of chemotherapy-induced nausea and vomiting after moderately emetogenic chemotherapy [Unpublished manuscript].2014:1-23.
59.
Abrams  DI, Hilton  JF, Leiser  RJ,  et al.  Short-term effects of cannabinoids in patients with HIV-1 infection: a randomized, placebo-controlled clinical trial.  Ann Intern Med. 2003;139(4):258-266.PubMedGoogle ScholarCrossref
60.
Timpone  JG, Wright  DJ, Li  N,  et al; Division of AIDS Treatment Research Initiative.  The safety and pharmacokinetics of single-agent and combination therapy with megestrol acetate and dronabinol for the treatment of HIV wasting syndrome: the DATRI 004 Study Group.  AIDS Res Hum Retroviruses. 1997;13(4):305-315.PubMedGoogle ScholarCrossref
61.
Struwe  M, Kaempfer  SH, Geiger  CJ,  et al.  Effect of dronabinol on nutritional status in HIV infection.  Ann Pharmacother. 1993;27(7-8):827-831.PubMedGoogle Scholar
62.
Beal  JE, Olson  R, Laubenstein  L,  et al.  Dronabinol as a treatment for anorexia associated with weight loss in patients with AIDS.  J Pain Symptom Manage. 1995;10(2):89-97.PubMedGoogle ScholarCrossref
63.
Ware  M, Fitzcharles  MA, Joseph  L, Shir  Y.  The effects of nabilone on sleep in fibromyalgia: results of a randomized controlled trial. Paper presented at: Canadian Rheumatology Association Meeting; February 18-21, 2009; Kananaskis, AB: Canada. Abstract 149  J Rheumatol. 2009;36(11):2607.Google Scholar
64.
Ware  MA, Fitzcharles  M-A, Joseph  L, Shir  Y.  The effects of nabilone on sleep in fibromyalgia: results of a randomized controlled trial.  Anesth Analg. 2010;110(2):604-610.PubMedGoogle ScholarCrossref
65.
Langford  RM, Mares  J, Novotna  A,  et al.  A double-blind, randomized, placebo-controlled, parallel-group study of THC/CBD oromucosal spray in combination with the existing treatment regimen, in the relief of central neuropathic pain in patients with multiple sclerosis.  J Neurol. 2013;260(4):984-997.PubMedGoogle ScholarCrossref
66.
Wilsey  B, Marcotte  T, Deutsch  R, Gouaux  B, Sakai  S, Donaghe  H.  Low-dose vaporized cannabis significantly improves neuropathic pain.  J Pain. 2013;14(2):136-148.PubMedGoogle ScholarCrossref
67.
Portenoy  RK, Ganae-Motan  ED, Allende  S,  et al.  Nabiximols for opioid-treated cancer patients with poorly-controlled chronic pain: a randomized, placebo-controlled, graded-dose trial.  J Pain. 2012;13(5):438-449.PubMedGoogle ScholarCrossref
68.
Ware  MA, Wang  T, Shapiro  S,  et al.  Smoked cannabis for chronic neuropathic pain: a randomized controlled trial.  CMAJ. 2010;182(14):E694-E701.PubMedGoogle ScholarCrossref
69.
Johnson  JR, Burnell-Nugent  M, Lossignol  D, Ganae-Motan  ED, Potts  R, Fallon  MT.  Multicenter, double-blind, randomized, placebo-controlled, parallel-group study of the efficacy, safety, and tolerability of THC:CBD extract and THC extract in patients with intractable cancer-related pain.  J Pain Symptom Manage. 2010;39(2):167-179.PubMedGoogle ScholarCrossref
70.
Selvarajah  D, Gandhi  R, Emery  CJ, Tesfaye  S.  Randomized placebo-controlled double-blind clinical trial of cannabis-based medicinal product (Sativex) in painful diabetic neuropathy: depression is a major confounding factor.  Diabetes Care. 2010;33(1):128-130.PubMedGoogle ScholarCrossref
71.
Ellis  RJ, Toperoff  W, Vaida  F,  et al.  Smoked medicinal cannabis for neuropathic pain in HIV: a randomized, crossover clinical trial.  Neuropsychopharmacology. 2009;34(3):672-680.PubMedGoogle ScholarCrossref
72.
Wilsey  B, Marcotte  T, Tsodikov  A,  et al.  A randomized, placebo-controlled, crossover trial of cannabis cigarettes in neuropathic pain.  J Pain. 2008;9(6):506-521.PubMedGoogle ScholarCrossref
73.
Narang  S, Gibson  D, Wasan  AD,  et al.  Efficacy of dronabinol as an adjuvant treatment for chronic pain patients on opioid therapy.  J Pain. 2008;9(3):254-264.PubMedGoogle ScholarCrossref
74.
Skrabek  RQ, Galimova  L, Ethans  K, Perry  D.  Nabilone for the treatment of pain in fibromyalgia.  J Pain. 2008;9(2):164-173.PubMedGoogle ScholarCrossref
75.
Frank  B, Serpell  MG, Hughes  J, Matthews  JNS, Kapur  D.  Comparison of analgesic effects and patient tolerability of nabilone and dihydrocodeine for chronic neuropathic pain: randomised, crossover, double blind study.  BMJ. 2008;336(7637):199-201.PubMedGoogle ScholarCrossref
76.
Nurmikko  TJ, Serpell  MG, Hoggart  B, Toomey  PJ, Morlion  BJ, Haines  D.  Sativex successfully treats neuropathic pain characterised by allodynia: a randomised, double-blind, placebo-controlled clinical trial.  Pain. 2007;133(1-3):210-220.PubMedGoogle ScholarCrossref
77.
Abrams  DI, Jay  CA, Shade  SB,  et al.  Cannabis in painful HIV-associated sensory neuropathy: a randomized placebo-controlled trial.  Neurology. 2007;68(7):515-521.PubMedGoogle ScholarCrossref
78.
Pinsger  M, Schimetta  W, Volc  D, Hiermann  E, Riederer  F, Pölz  W.  [Benefits of an add-on treatment with the synthetic cannabinomimetic nabilone on patients with chronic pain—a randomized controlled trial].  Wien Klin Wochenschr. 2006;118(11-12):327-335.PubMedGoogle ScholarCrossref
79.
Blake  DR, Robson  P, Ho  M, Jubb  RW, McCabe  CS.  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). 2006;45(1):50-52.PubMedGoogle ScholarCrossref
80.
Rog  DJ, Nurmikko  TJ, Friede  T, Young  CA.  Randomized, controlled trial of cannabis-based medicine in central pain in multiple sclerosis.  Neurology. 2005;65(6):812-819.PubMedGoogle ScholarCrossref
81.
Berman  JS, Symonds  C, Birch  R.  Efficacy of two cannabis based medicinal extracts for relief of central neuropathic pain from brachial plexus avulsion: results of a randomised controlled trial.  Pain. 2004;112(3):299-306.PubMedGoogle ScholarCrossref
82.
Svendsen  KB, Jensen  TS, Bach  FW.  Does the cannabinoid dronabinol reduce central pain in multiple sclerosis? randomised double blind placebo controlled crossover trial.  BMJ. 2004;329(7460):253.PubMedGoogle ScholarCrossref
83.
Karst  M, Salim  K, Burstein  S, Conrad  I, Hoy  L, Schneider  U.  Analgesic effect of the synthetic cannabinoid CT-3 on chronic neuropathic pain: a randomized controlled trial.  JAMA. 2003;290(13):1757-1762.PubMedGoogle ScholarCrossref
84.
Noyes  R  Jr, Brunk  SF, Baram  DA, Canter  A.  Analgesic effect of delta-9-tetrahydrocannabinol.  J Clin Pharmacol. 1975;15(2-3):139-143.PubMedGoogle ScholarCrossref
85.
Lynch  ME, Cesar-Rittenberg  P, Hohmann  AG.  A double-blind, placebo-controlled, crossover pilot trial with extension using an oral mucosal cannabinoid extract for treatment of chemotherapy-induced neuropathic pain.  J Pain Symptom Manage. 2014;47(1):166-173.PubMedGoogle ScholarCrossref
86.
Wallace  M, Atkinson  J, Gouaux  B, Marcotte  T, Umlauf  A.  Effect of smoked cannabis on painful diabetic peripheral neuropathy. Paper presented at: 32nd Annual Scientific Meeting of the American Pain Society; May 9-11, 2013; New Orleans: LA.  J Pain. 2013;14(4)(suppl 1):S62 doi:10.1016/j.jpain.2013.01.587.Google ScholarCrossref
87.
Berman  J, Bosworth  T, Guy  G, Stott  C; Sativex Spinal Cord Injury Study Group.  Sativex in the treatment of central neuropathic pain due to spinal cord injury: a randomised controlled study. Paper presented at: British Pain Society Annual Scientific Meeting; April 2007; Glasgow: United Kingdom.
88.
Serpell  M, Ratcliffe  S, Hovorka  J,  et al.  A double-blind, randomized, placebo-controlled, parallel group study of THC/CBD spray in peripheral neuropathic pain treatment.  Eur J Pain. 2014;18(7):999-1012.PubMedGoogle ScholarCrossref
89.
Fitzcharles  MA, Shir  Y, Joseph  L, Ware  MA.  The effects of nabilone on insomnia in fibromyalgia: results of a randomized controlled trial. Paper presented at: American College of Rheumatology/Association of Rheumatology Health Professionals Annual Scientific Meeting (ACR/ARHP 09); November 6-11, 2009; Atlanta: GA.  Arthritis Rheum. 2009;60:1429.Google Scholar
90.
McGill University Health Center.  Nabilone versus amitriptyline in improving quality of sleep in patients with fibromyalgia. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00381199 Accessed April 7, 2014.
91.
GW Pharmaceuticals Ltd.  Sativex versus placebo when added to existing treatment for central neuropathic pain in MS.http://ClinicalTrials.gov/show/NCT00391079 Accessed April 7, 2014.
92.
Svendsen  KB, Jensen  TS, Bach  FW.  [Effect of the synthetic cannabinoid dronabinol on central pain in patients with multiple sclerosis–secondary publication].  Ugeskr Laeger.2005;167(25-31):2772-2774.PubMedGoogle Scholar
93.
Salim  K, Schneider  U, Burstein  S, Hoy  L, Karst  M.  Pain measurements and side effect profile of the novel cannabinoid ajulemic acid.  Neuropharmacology. 2005;48(8):1164-1171.PubMedGoogle ScholarCrossref
94.
Pinsger  M.  Benefit of an add-on-treatment with a synthetic cannabinomimeticum on patients with chronic back pain-a randomized controlled trial. Paper presented at 8th International Conference on Early Psychosis: From Neurobiology to Public Policy; October 11-13, 2012; San Francisco: CA.  Eur Spine J. 2012;21(11):2366 doi:10.1007/s00586-012-2522-6.Google Scholar
95.
Nurmikko  TJ, Serpell  MG, Hoggart  B, Toomey  PJ, Morlion  BJ.  A multi-centre, double-blind, randomized, controlled trial of oro-mucosal cannabis based medicine in the treatment of neuropathic pain characterized by allodynia.  Neurology. 2005;64(suppl 1):A374.Google ScholarCrossref
96.
Issa  MA, Narang  S, Jamison  RN,  et al.  The subjective psychoactive effects of oral dronabinol studied in a randomized, controlled crossover clinical trial for pain.  Clin J Pain. 2014;30(6):472-478.PubMedGoogle ScholarCrossref
97.
Abrams  DI, Jay  CA, Vizoso  H,  et al.  Smoked cannabis therapy for HIV-related painful peripheral neuropathy: results of a randomized, placebo-controlled clinical trial. Paper presented at: IACM 3rd Conference on Cannabinoids in Medicine; September 9-10, 2005; Leiden, the Netherlands.
98.
Young  CA, Rog  DJ.  Randomised controlled trial of cannabis based medicinal extracts (CBME) in central neuropathic pain due to multiple sclerosis. Paper presented at: IV Congress of the European Federation of IASP Chapters (EFIC); September 2-6, 2003; Prague, Czech Republic.
99.
Berman  J, Lee  J, Cooper  M,  et al.  Efficacy of two cannabis-based medicinal extracts for relief of central neuropathic pain from brachial plexus avulsion: results of a randomised controlled trial. Paper presented at: Pain Society Annual Meeting; April 1-4, 2003; Glasgow, United Kingdom.  Anaesthesia. 2003;58(9):938 doi:10.1046/j.1365-2044.2003.03408_3.x.Google ScholarCrossref
100.
Center for Medicinal Cannabis Research.  Effects of smoked marijuana on neuropathic pain. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00254761. Accessed April 7, 2014.
101.
Center for Medicinal Cannabis Research.  Medicinal cannabis for painful HIV neuropathy. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00255580. Accessed April 7, 2014.
102.
University of California Davis.  Center for Medicinal Cannabis Research, VA Northern California Health Care System. Effects of vaporized marijuana on neuropathic pain. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT01037088. Accessed April 7, 2014.
103.
Center for Medicinal Cannabis Research.  Marijuana for HIV-related peripheral neuropathy. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00046722. Accessed April 7, 2014.
104.
GW Pharmaceuticals Ltd.  A study of Sativex® for pain relief in patients with advanced malignancy. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00530764. Accessed April 7, 2014.
105.
GW Pharmaceuticals Ltd.  A study of sativex® for pain relief in patients with advanced malignancy. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00674609. Accessed April 7, 2014.
106.
GW Pharmaceuticals Ltd.  A study of sativex® for relief of peripheral neuropathic pain associated with allodynia. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00711880. Accessed April 7, 2014.
107.
GW Pharmaceuticals Ltd.  A study of sativex in the treatment of central neuropathic pain due to multiple sclerosis. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT01604265. Accessed April 7, 2014.
108.
GW Pharmaceuticals Ltd.  A study of sativex® for pain relief due to diabetic neuropathy. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00710424. Accessed April 7, 2014.
109.
GW Pharmaceuticals Ltd.  A study of Sativex® for pain relief of peripheral neuropathic pain, associated with allodynia. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00710554. Accessed April 7, 2014.
110.
Mary Lynch, Capital District Health Authority Canada.  Sativex for treatment of chemotherapy induced neuropathic pain. .ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00872144. Accessed April 7, 2014.
111.
Brigham and Women’s Hospital; Solvay Pharmaceuticals.  Study to evaluate the efficacy of dronabinol (Marinol) as add-on therapy for patients on opioids for chronic pain. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00153192. Accessed April 7, 2014.
112.
Winnipeg Regional Health Authority; Valeant Canada Limited.  A trial assessing the effect of nabilone on pain and quality of life in patients with fibromyalgia. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00272207. Accessed April 7, 2014.
113.
GW Pharma Ltd.  A double blind, randomised, placebo controlled parallel group study of cannabis based medicine extract (CBME), in the treatment of peripheral neuropathic pain characterised by allodynia. metaRegister of Controlled Trials. http://www.controlled-trials.com/ISRCTN38250575. Accessed April 7, 2014.
114.
Montreal General Hospital.  Pilot study of smoked cannabis for chronic neuropathic pain. metaRegister of Controlled Trials (mRCT), http://www.controlled-trials.com/ISRCTN68314063. Accessed April 7, 2014.
115.
GW Pharma Ltd.  A double blind, randomised, placebo controlled, parallel group study of Sativex, in the treatment of subjects with peripheral neuropathic pain associated with allodynia. EU Clinical Trials Register. https://www.clinicaltrialsregister.eu/ctr-search/search?query=eudract_number:2004-002531-32. Accessed April 8, 2014.
116.
Cambridge Laboratories Ltd.  A randomised, crossover, double blind comparison of the analgesic effect and patient tolerability of nabilone and dihydrocodeine in chronic neuropathic pain. metaRegister of Controlled Trials. http://isrctn.org/ISRCTN15330757. Accessed April 7, 2014.
117.
Selvarajah  D, Gandhi  RA, Witte  D, Bowler  H, Emery  C, Tesfaye  S.  Treatment of painful diabetic neuropathy with Sativex (a cannabis based medicinal product)—results of a randomised placebo controlled trial.  Diabetologia. 2006;49 (suppl 1):671-672 doi:10.1007/s00125-006-0358-5.Google Scholar
118.
Rog  DJ, Nurmikko  T, Young  C, Sarantis  NS.  Randomized controlled trial of sativex, a cannabis based medicine (CBM), in central neuropathic pain due to multiple sclerosis, followed by an open-label extension.  Neurology. 2006;66(5):A31.Google Scholar
119.
Ventegodt  S, Merrick  J.  Psychoactive drugs and quality of life.  ScientificWorldJournal. 2003;3:694-706.PubMedGoogle ScholarCrossref
120.
Wallace  MS, Marcotte  TD, Umlauf  A, Gouaux  B, Atkinson  JH.  Efficacy of inhaled cannabis on painful diabetic neuropathy. J Pain.2015;pii:S1526-5900(1515)00601-X.PubMed
121.
Turcotte  D, Doupe  M, Torabi  M,  et al.  Nabilone as an adjunctive to gabapentin for multiple sclerosis-induced neuropathic pain: a randomized controlled trial.  Pain Med. 2015;16(1):149-159.PubMedGoogle ScholarCrossref
122.
Zajicek  JP, Sanders  HP, Wright  DE,  et al.  Cannabinoids in multiple sclerosis (CAMS) study: safety and efficacy data for 12 months follow up.  J Neurol Neurosurg Psychiatry. 2005;76(12):1664-1669.PubMedGoogle ScholarCrossref
123.
Zajicek  JP, Hobart  JC, Slade  A, Barnes  D, Mattison  PG; MUSEC Research Group.  Multiple sclerosis and extract of cannabis: results of the MUSEC trial.  J Neurol Neurosurg Psychiatry. 2012;83(11):1125-1132.PubMedGoogle ScholarCrossref
124.
Corey-Bloom  J, Wolfson  T, Gamst  A,  et al.  Smoked cannabis for spasticity in multiple sclerosis: a randomized, placebo-controlled trial.  CMAJ. 2012;184(10):1143-1150.PubMedGoogle ScholarCrossref
125.
Collin  C, Ehler  E, Waberzinek  G,  et al.  A double-blind, randomized, placebo-controlled, parallel-group study of Sativex, in subjects with symptoms of spasticity due to multiple sclerosis.  Neurol Res. 2010;32(5):451-459.PubMedGoogle ScholarCrossref
126.
Pooyania  S, Ethans  K, Szturm  T, Casey  A, Perry  D.  A randomized, double-blinded, crossover pilot study assessing the effect of nabilone on spasticity in persons with spinal cord injury.  Arch Phys Med Rehabil. 2010;91(5):703-707.PubMedGoogle ScholarCrossref
127.
Collin  C, Davies  P, Mutiboko  IK, Ratcliffe  S; Sativex Spasticity in MS Study Group.  Randomized controlled trial of cannabis-based medicine in spasticity caused by multiple sclerosis.  Eur J Neurol. 2007;14(3):290-296.PubMedGoogle ScholarCrossref
128.
Freeman  RM, Adekanmi  O, Waterfield  MR, Waterfield  AE, Wright  D, Zajicek  J.  The effect of cannabis on urge incontinence in patients with multiple sclerosis: a multicentre, randomised placebo-controlled trial (CAMS-LUTS).  Int Urogynecol J Pelvic Floor Dysfunct. 2006;17(6):636-641.PubMedGoogle ScholarCrossref
129.
Wade  DT, Makela  P, Robson  P, House  H, Bateman  C.  Do cannabis-based medicinal extracts have general or specific effects on symptoms in multiple sclerosis? a double-blind, randomized, placebo-controlled study on 160 patients.  Mult Scler. 2004;10(4):434-441.PubMedGoogle ScholarCrossref
130.
Vaney  C, Heinzel-Gutenbrunner  M, Jobin  P,  et al.  Efficacy, safety and tolerability of an orally administered cannabis extract in the treatment of spasticity in patients with multiple sclerosis: a randomized, double-blind, placebo-controlled, crossover study.  Mult Scler. 2004;10(4):417-424.PubMedGoogle ScholarCrossref
131.
Zajicek  J, Fox  P, Sanders  H,  et al; UK MS Research Group.  Cannabinoids for treatment of spasticity and other symptoms related to multiple sclerosis (CAMS study): multicentre randomised placebo-controlled trial.  Lancet. 2003;362(9395):1517-1526.PubMedGoogle ScholarCrossref
132.
Killestein  J, Hoogervorst  ELJ, Reif  M,  et al.  Safety, tolerability, and efficacy of orally administered cannabinoids in MS.  Neurology. 2002;58(9):1404-1407.PubMedGoogle ScholarCrossref
133.
Hobart  JC, Zajicek  JP.  Cannabis as a symptomatic treatment for MS: Clinically meaningful MUSEC to the stiffness and walking problems of people with MS. Paper presented at: 28th Congress of the European Committee for Treatment and Research in Multiple Sclerosis; October 10-13, 2012; Lyon: France.  Mult Scler. 2012;18(4 suppl 1):247.PubMedGoogle Scholar
134.
Zajicek  J, Reif  M, Schnelle  M.  Cannabis extract in the treatment of muscle stiffness and other symptoms in multiple sclerosis—Results of the MUSEC study. Paper presented at: 25th Congress of the European Committee for Treatment and Research in Multiple Sclerosis; September 9-12, 2009; Dusseldorf: Germany.  Mult Scler. 2009;15(9)(suppl S):S274 doi:10.1177/1352458509107025.Google Scholar
135.
Killestein  J, Hoogervorst  ELJ, Kalkers  NF,  et al.  The effects of orally administred cannabinoids in multiple sclerosis patients: a pilot study.  Mult Scler. 2000;6(1 suppl 1):S28 doi:10.1177/135245850000600101.Google Scholar
136.
Zajicek  J, Reif  M, Schnelle  M; UK MUSEC Study Investigators.  Cannabis extract in the treatment of muscle stiffness and other symptoms in multiple sclerosis – results of the MUSEC study. Paper presented at: IACM 5th Conference on Cannabinoids in Medicine; October 2-3, 2009; Cologne, Germany.
137.
Collin  C, Ambler  Z, Kent  R, McCalla  R.  A randomised controlled study of Sativex® in patients with symptoms of spasticity due to multiple sclerosis. Paper presented at: 22nd Congress of the ECTRIMS; September 27-30, 2006; Madrid, Spain.
138.
Robson  P, Wade  D, Makela  P, House  H, Bateman  C.  Cannabis-based medicinal extract (Sativex) produced significant improvements in a subjective measure of spasticity which were maintained on long-term treatment with no evidence of tolerance. Paper presented at: IACM 3rd Conference on Cannabinoids in Medicine; September 9-10, 2005; Leiden, the Netherlands.
139.
Center for Medicinal Cannabis Research.  Short-term effects of medicinal cannabis therapy on spasticity in multiple sclerosis. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00248378. Accessed April 7, 2014.
140.
Institut fur Klinische Forschung Germany; Weleda AG. Multiple Sclerosis and Extract of Cannabis (MUSEC) study. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00552604. Accessed April 7, 2014.
141.
GW Pharmaceuticals Ltd.  A study of Sativex® for relief of spasticity in subjects with multiple sclerosis. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00711646. Accessed April 7, 2014.
142.
GW Pharmaceuticals Ltd.  A study to evaluate the efficacy of Sativex in relieving symptoms of spasticity due to multiple sclerosis. ClinicalTrials.gov.http://ClinicalTrials.gov/show/NCT01599234. Accessed April 7, 2014.
143.
GW Pharmaceuticals Ltd.  An investigation of delta-9-tetrahydrocannabinol (THC) and cannabidiol (CBD) in multiple sclerosis patients. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT01610700. Accessed April 7, 2014.
144.
University of Manitoba, Valeant Canada Limited.  Randomized double blind cross over study for nabilone in spasticity in spinal cord injury persons. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00623376. Accessed April 7, 2014.
145.
Medical Research Council (MRC).  A multiple randomised controlled trial of cannabinoids on spasticity in multiple sclerosis (MS). metaRegister of Controlled Trials. http://www.controlled-trials.com/ISRCTN39371386. Accessed April 7, 2014.
146.
Gesellschaft fuer klinische Forschung e.V.  Multiple Sclerosis and Extract of Cannabis (MUSEC): a randomised, double-blind, placebo-controlled phase III trial to determine the efficacy and safety of a standardised oral extract of cannabis sativa for the symptomatic relief of muscle stiffness and pain in multiple sclerosis. EU Clinical Trials Register. https://www.clinicaltrialsregister.eu/ctr-search/search?query=eudract_number:2005-005263-29. Accessed April 8, 2014.
147.
Corey-Bloom  J, Wolfson  TJ, Anthony  GC, Bentley  H, Gouaux  B.  Short-term effects off medicinal cannabis on spasticity in multiple sclerosis.  Neurology. 2008;70(11)(suppl 1):A86-A87.Google Scholar
148.
Leocani  L, Nuara  A, Houdayer  E,  et al.  Effect of THC-CBD oromucosal spray (Sativex) on measures of spasticity in multiple sclerosis: a double-blind, placebo-controlled, crossover study. Paper presented at: Joint Americas Committee for Treatment and Research in Multiple Sclerosis ACTRIMS—European Committee for Treatment and Research in Multiple Sclerosis ECTRIMS Meeting; September 10-13, 2014; Boston, MA.  Mult Scler. 2014;20(1 suppl 1):498 doi:10.1177/1352458514547846.Google Scholar
149.
Van Amerongen  G, Beumer  T, Killestein  J, Groeneveld  GJ.  Individualized dosing of a novel oral DELTA9-THC formulation improves subjective spasticity and pain in patients with progressive multiple sclerosis. Paper presented at Joint Americas Committee for Treatment and Research in Multiple Sclerosis ACTRIMS—European Committee for Treatment and Research in Multiple Sclerosis ECTRIMS Meeting; September 10-13, 2014; Boston,MA.  Mult Scler. 2014;20(1)(suppl 1):478-479 doi:10.1177/1352458514547846.Google Scholar
150.
Bergamaschi  MM, Queiroz  RH, Chagas  MH,  et al.  Cannabidiol reduces the anxiety induced by simulated public speaking in treatment-naïve social phobia patients.  Neuropsychopharmacology. 2011;36(6):1219-1226.PubMedGoogle ScholarCrossref
151.
Leweke  FM, Piomelli  D, Pahlisch  F,  et al.  Cannabidiol enhances anandamide signaling and alleviates psychotic symptoms of schizophrenia.  Transl Psychiatry. 2012;2:e94.PubMedGoogle ScholarCrossref
152.
Leweke  FM, Gerth  CW, Nolden  BM,  et al.  Cannabidiol as antipsychotic. Paper presented at: 21st ECNP Congress; August 30, 2008; Barcelona, Spain.  Eur Neuropsychopharmacol. 2008;18(S4):S171 doi:10.1016/S0924-977X(08)70156-1.Google ScholarCrossref
153.
Leweke  FM, Koethe  D, Pahlisch  F,  et al.  Antipsychotic effects of cannabidiol. Paper presented at: 17th European Psychiatric Association, EPA Congress; January 24-28, 2009; Lisbon, Portugal.  Eur Psychiatry. 2009;24(supp 1):S207 doi:10.1016/S0924-9338(09)70440-7.Google ScholarCrossref
154.
University of Cologne.  Evaluation of the antipsychotic efficacy of cannabidiol in acute schizophrenic psychosis. ClinicalTrials.gov. http://ClinicalTrials.gov/show/NCT00628290. Accessed April 7, 2014.
155.
Rohleder  C, Pahlisch  F, Schaefer  C,  et al.  The endocannabinoid system as a pharmacological target for antipsychotic treatment and more? Paper presented at: 8th International Conference on Early Psychosis: From Neurobiology to Public Policy; October 11-13, 2012; San Francisco, CA.  Early Interv Psychiatry. 2012;6(suppl 1):7 doi:10.1111/j.1751-7893.2012.00392.x.Google Scholar
156.
Markus  F, Leweke  M, Kranaster  L,  et al.  The efficacy of cannabidiol in the treatment of schizophrenia—A translational approach. Paper presented at: 13th International Congress on Schizophrenia Research, ICOSR; April 2-6, 2011; Colorado Springs, CO.  Schizophr Bull. 2011;37(suppl 1):313 doi:10.1093/schbul/sbq173.Google Scholar
157.
Leweke  FM, Kranaster  L, Hellmich  M, Koethe  D.  Cannabidiol as a new type of an antipsychotic: results from a placebo-controlled clinical trial. Paper presented at: 49th Annual Conference of the American College of Neuropsychopharmacology, ACNP 2010; December 5-9, 2010; Miami Beach, FL.  Neuropsychopharmacology. 2010;35(suppl 1):S280 doi:10.1038/npp.2010.217.Google Scholar
158.
Leweke  FM, Hellmich  M, Kranaster  L, Koethe  D.  Cannabidiol as a new type of an antipsychotic: results from a placebo-controlled clinical trial. Paper presented at: 67th Annual Scientific Convention and Meeting of the Society of Biological Psychiatry; May 3-5, 2012; Philadelphia, PA. Biol Psychiatry. 2012;78(8)(suppl 1):63S.
159.
Tomida  I, Azuara-Blanco  A, House  H, Flint  M, Pertwee  RG, Robson  PJ.  Effect of sublingual application of cannabinoids on intraocular pressure: a pilot study.  J Glaucoma. 2006;15(5):349-353.PubMedGoogle ScholarCrossref
160.
Müller-Vahl  KR, Prevedel  H, Theloe  K, Kolbe  H, Emrich  HM, Schneider  U.  Treatment of Tourette syndrome with delta-9-tetrahydrocannabinol (delta 9-THC): no influence on neuropsychological performance.  Neuropsychopharmacology. 2003;28(2):384-388.PubMedGoogle ScholarCrossref
161.
Müller-Vahl  KR, Schneider  U, Prevedel  H,  et al.  Delta 9-tetrahydrocannabinol (THC) is effective in the treatment of tics in Tourette syndrome: a 6-week randomized trial.  J Clin Psychiatry. 2003;64(4):459-465.PubMedGoogle ScholarCrossref
162.
Müller-Vahl  KR, Koblenz  A, Jöbges  M, Kolbe  H, Emrich  HM, Schneider  U.  Influence of treatment of Tourette syndrome with delta9-tetrahydrocannabinol (delta9-THC) on neuropsychological performance.  Pharmacopsychiatry. 2001;34(1):19-24.PubMedGoogle ScholarCrossref
163.
Müller-Vahl  KR, Schneider  U, Koblenz  A,  et al.  Treatment of Tourette’s syndrome with Delta 9-tetrahydrocannabinol (THC): a randomized crossover trial.  Pharmacopsychiatry. 2002;35(2):57-61.PubMedGoogle ScholarCrossref
164.
International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use.  MeDRA (Medical Dictionary for Regulatory Activities).http://www.meddra.org/. Accessed September 2, 2014.
165.
McGowan  J, Sampson  M, Lefebvre  C.  An evidence based checklist for the peer review of electronic search strategies (PRESS EBC).  Evid Based Libr Inf Pract. 2010;5(1):1-6. http://ejournals.library.ualberta.ca/index.php/EBLIP/article/view/7402. Accessed Month, date, year.Google Scholar
166.
Elbourne  DR, Altman  DG, Higgins  JP, Curtin  F, Worthington  HV, Vail  A.  Meta-analyses involving cross-over trials: methodological issues.  Int J Epidemiol. 2002;31(1):140-149.PubMedGoogle ScholarCrossref
167.
Lakhan  SE, Rowland  M.  Whole plant cannabis extracts in the treatment of spasticity in multiple sclerosis: a systematic review.  BMC Neurol. 2009;9:59.PubMedGoogle ScholarCrossref
168.
Sevilla Guerra  S.  Are cannabinoids more effective than placebo in decreasing MS-related bladder dysfunction?  Br J Neurosci Nurs. 2012;8(2):71-78 doi:10.12968/bjnn.2012.8.2.71.Google ScholarCrossref
169.
Shakespeare  D, Boggild  M, Young  CA.  Anti-spasticity agents for multiple sclerosis.  Cochrane Database Syst Rev. 2003;(4):CD001332.PubMedGoogle Scholar
170.
Wade  DT, Collin  C, Stott  C, Duncombe  P.  Meta-analysis of the efficacy and safety of Sativex (nabiximols), on spasticity in people with multiple sclerosis.  Mult Scler. 2010;16(6):707-714.PubMedGoogle ScholarCrossref
171.
Brook  JS, Lee  JY, Finch  SJ, Brown  EN.  Course of comorbidity of tobacco and marijuana use: psychosocial risk factors.  Nicotine Tob Res. 2010;12(5):474-482.PubMedGoogle ScholarCrossref
172.
Machado Rocha  FC, Stéfano  SC, De Cássia Haiek  R, Rosa Oliveira  LM, Da Silveira  DX.  Therapeutic use of Cannabis sativa on chemotherapy-induced nausea and vomiting among cancer patients: systematic review and meta-analysis.  Eur J Cancer Care (Engl). 2008;17(5):431-443.PubMedGoogle ScholarCrossref
173.
Phillips  RS, Gopaul  S, Gibson  F,  et al Antiemetic medication for prevention and treatment of chemotherapy induced nausea and vomiting in childhood.  Cochrane Database Syst Rev. 2010;(9):CD007786.PubMedGoogle Scholar
174.
Tramèr  MR, Carroll  D, Campbell  FA, Reynolds  DJ, Moore  RA, McQuay  HJ.  Cannabinoids for control of chemotherapy induced nausea and vomiting: quantitative systematic review.  BMJ. 2001;323(7303):16-21.PubMedGoogle ScholarCrossref
175.
van den Elsen  GAH, Ahmed  AIA, Lammers  M,  et al.  Efficacy and safety of medical cannabinoids in older subjects: a systematic review.  Ageing Res Rev. 2014;14(1):56-64.PubMedGoogle ScholarCrossref
176.
Burns  TL, Ineck  JR.  Cannabinoid analgesia as a potential new therapeutic option in the treatment of chronic pain.  Ann Pharmacother. 2006;40(2):251-260.PubMedGoogle ScholarCrossref
177.
Campbell  FA, Tramèr  MR, Carroll  D, Reynolds  DJ, Moore  RA, McQuay  HJ.  Are cannabinoids an effective and safe treatment option in the management of pain? a qualitative systematic review.  BMJ. 2001;323(7303):13-16.PubMedGoogle ScholarCrossref
178.
Canadian Agency for Drugs and Technologies in Health (CADTH).  Cannabinoids as Co-Analgesics: Review of Clinical Effectiveness. Ottawa, Canada: Canadian Agency for Drugs and Technologies in Health;2010.
179.
Canadian Agency for Drugs and Technologies in Health (CADTH).  Cannabinoids for the Management of Neuropathic Pain: Review of Clinical Effectiveness. Ottawa, Canada: Canadian Agency for Drugs and Technologies in Health);2010.
180.
Davis  MP.  Oral nabilone capsules in the treatment of chemotherapy-induced nausea and vomiting and pain.  Expert Opin Investig Drugs. 2008;17(1):85-95.PubMedGoogle ScholarCrossref
181.
Iskedjian  M, Bereza  B, Gordon  A, Piwko  C, Einarson  TR.  Meta-analysis of cannabis based treatments for neuropathic and multiple sclerosis-related pain.  Curr Med Res Opin. 2007;23(1):17-24.PubMedGoogle ScholarCrossref
182.
Jawahar  R, Oh  U, Yang  S, Lapane  KL.  A systematic review of pharmacological pain management in multiple sclerosis.  Drugs. 2013;73(15):1711-1722.PubMedGoogle ScholarCrossref
183.
Kung  T, Hochman  J, Sun  Y,  et al.  Efficacy and safety of cannabinoids for pain in musculoskeletal diseases: a systematic review and meta-analysis. Paper presented at: 2nd Mexican-Canadian Congress of Rheumatology; February 10-15, 2011; Cancun, Mexico.  J Rheumatol. 2011;38(6):1171 doi:10.3899/jrheum.110506.Google Scholar
184.
Lynch  ME, Campbell  F.  Cannabinoids for treatment of chronic non-cancer pain; a systematic review of randomized trials.  Br J Clin Pharmacol. 2011;72(5):735-744.PubMedGoogle ScholarCrossref
185.
Martín-Sánchez  E, Furukawa  TA, Taylor  J, Martin  JL.  Systematic review and meta-analysis of cannabis treatment for chronic pain.  Pain Med. 2009;10(8):1353-1368.PubMedGoogle ScholarCrossref
186.
Phillips  TJ, Cherry  CL, Cox  S, Marshall  SJ, Rice  AS.  Pharmacological treatment of painful HIV-associated sensory neuropathy: a systematic review and meta-analysis of randomised controlled trials.  PLoS One. 2010;5(12):e14433.PubMedGoogle ScholarCrossref
187.
Pittler  MH, Ernst  E.  Complementary therapies for neuropathic and neuralgic pain: systematic review.  Clin J Pain. 2008;24(8):731-733.PubMedGoogle ScholarCrossref
188.
Richards  BL, Whittle  SL, Buchbinder  R.  Neuromodulators for pain management in rheumatoid arthritis.  Cochrane Database Syst Rev. 2012;(1):CD008921.PubMedGoogle Scholar
189.
Boychuk  DG, Goddard  G, Mauro  G, Orellana  MF.  The effectiveness of cannabinoids in the management of chronic nonmalignant neuropathic pain: a systematic review.  J Oral Facial Pain Headache. 2015;29(1):7-14.PubMedGoogle ScholarCrossref
190.
Parsai  S, Herman  R, Johnson  S.  Systematic literature review of randomized controlled trials to evaluate the efficacy of medical marijuana for analgesia. Paper presented at: Annual Meeting of the American College of Clinical Pharmacy; October 12-15, 2014; Austin, TX.  Pharmacotherapy. 2014;34(10):e287 doi:10.1002/phar.1497.Google Scholar
191.
Lynch  ME, Ware  MA.  Cannabinoids for the treatment of chronic non-cancer pain: an updated systematic review of randomized controlled trials [published online March 22, 2015].  J Neuroimmune Pharmacol doi.org/10.1007/s11481-015-9600-6.PubMedGoogle Scholar
192.
Rathbone  J, Variend  H, Mehta  H. Cannabis and schizophrenia.  Cochrane Database Syst Rev.2008;(3):CD004837.PubMedGoogle Scholar
193.
Schoeler  T, Kambeitz  J, Bhattacharyya  S.  The effect of cannabis on memory function in users with and without a psychotic disorder: a meta-analysis. Paper presented at: 26th European College of Neuropsychopharmacology, ECNP Congress; October 5-9, 2013; Barcelona, Spain.  Eur Neuropsychopharmacol. 2013;23:S216-S217 doi:10.1016/S0924-977X(13)70334-1.Google ScholarCrossref
194.
Zammit  S, Moore  THM, Lingford-Hughes  A,  et al.  Effects of cannabis use on outcomes of psychotic disorders: systematic review.  Br J Psychiatry. 2008;193(5):357-363.PubMedGoogle ScholarCrossref
195.
Curtis  A, Clarke  CE, Rickards  HE.  Cannabinoids for Tourette’s syndrome.  Cochrane Database Syst Rev. 2009;CD0065652009(4):CD006565.PubMedGoogle Scholar
196.
Waldon  K, Hill  J, Termine  C, Balottin  U, Cavanna  AE.  Trials of pharmacological interventions for Tourette syndrome: a systematic review.  Behav Neurol. 2013;26(4):265-273.PubMedGoogle ScholarCrossref
197.
Boers  M.  Updated Consolidated Standards of Reporting Trials (CONSORT): it just gets better.  J Clin Epidemiol. 2010;63(8):813-814.PubMedGoogle ScholarCrossref
×