Scolaris Content Display Scolaris Content Display

Ubat berasaskan ganja untuk kesakitan neuropatik kronik pada orang dewasa

Contraer todo Desplegar todo

Referencias

Bermann 2004 {published data only}

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;112:299-306. CENTRAL

Ellis 2009 {published data only}

Ellis RJ, Toperoff W, Vaida F, Van den Brande G, Gonzales J, Gouaux B, et al. Smoked medicinal cannabis for neuropathic pain in HIV: a randomized, crossover clinical trial. Neuropsychopharmacology 2009;34(3):672-80. CENTRAL

Frank 2008 {published data only}

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

Langford 2013 {published data only}

Langford RM, Mares J, Novotna A, Vachova M, Novakova I, Notcutt W, et al. A double-blind, randomized, placebo-controlled, parallel-group study of THC/CBD oromucosal spray in combination with the existing treatment regimen, in the relief of central neuropathic pain in patients with multiple sclerosis. Journal of Neurology 2013;260:984-97. CENTRAL

Lynch 2014 {published data only}

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. Journal of Pain and Symptom Management 2014;47:166-73. CENTRAL

NCT00710424 {published data only}

NCT 00710424. A study of Sativex® for pain relief due to diabetic neuropathy. clinicaltrials.gov/ct2/results?term=NCT00710424+&Search=Search (first Posted 4 July 2008). CENTRAL

NCT01606176 {published data only}

NCT01606176. A Study to Evaluate the Effects of Cannabis Based Medicine in Patients With Pain of Neurological Origin. clinicaltrials.gov/ct2/results?cond=&term=NCT01606176&cntry1=&state1=&recrs= (first posted 25 May 2012). CENTRAL

NCT01606202 {published data only}

NCT 01606202. A study of cannabis based medicine extracts and placebo in patients with pain due to spinal cord injury. clinicaltrials.gov/ct2/results?term= NCT 01606202&Search=Search (first posted 25 May 2012). CENTRAL

Nurmikko 2007 {published data only}

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;133:210-20. CENTRAL

Rog 2005 {published data only}

Rog DJ, Nurmikko TJ, Friede T, Young CA. Randomized, controlled trial of cannabis-based medicine in central pain in multiple sclerosis. Neurology 2005;65:812-9. CENTRAL

Schimrigk 2017 {published data only}

Schimrigk S, Marziniak M, Neubauer C, Kugler EM, Werner G, Abramov-Sommariva D. Dronabinol is a safe long-term treatment option for neuropathic pain patients. European Neurology 2017;78:320-9. CENTRAL

Selvarajah 2010 {published data only}

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;33:128-30. CENTRAL

Serpell 2014 {published data only}

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

Svendsen 2004 {published data only}

Svendsen KB, Jensen TS, Bach FW. Does the cannabinoid dronabinol reduce central pain in multiple sclerosis? Randomised double blind placebo controlled crossover trial. BMJ 2004;329:253. CENTRAL

Toth 2012 {published data only}

Toth C, Mawani S, Brady S, Chan C, Liu C, Mehina E, et al. An enriched-enrolment, randomized withdrawal, flexible-dose, double-blind, placebo-controlled, parallel assignment efficacy study of nabilone as adjuvant in the treatment of diabetic peripheral neuropathic pain. Pain 2012;153:2073-82. CENTRAL

Ware 2010 {published data only}

Ware MA, Wang T, Shapiro S, Robinson A, Ducruet T, Huynh T, et al. Smoked cannabis for chronic neuropathic pain: a randomized controlled trial. Canadian Medical Association Journal 2010;184:E694-701. CENTRAL

Abrams 2007 {published data only}

Abrams DI, Jay CA, Shade SB, Vizoso H, Reda H, Press S, et al. Cannabis in painful HIV-associated sensory neuropathy: a randomized placebo-controlled trial. Neurology 2007;68:515-21. CENTRAL

Corey‐Bloom 2012 {published data only}

Corey-Bloom J, Wolfson T, Gamst A, Jin S, Marcotte TD, Bentley H, et al. Smoked cannabis for spasticity in multiple sclerosis: a randomized, placebo-controlled trial. Canadian Medical Association Journal 2012;184:1143-50. CENTRAL

Karst 2003 {published data only}

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

Notcutt 2011 {published data only}

Notcutt W, Price M, Miller R, Newport S, Phillips C, Simmons S, et al. Initial experiences with medicinal extracts of cannabis for chronic pain: results from 34 'N of 1' studies. Anaesthesia 2004;59:440-52. CENTRAL

Novotna 2011 {published data only}

Novotna A, Mares J, Ratcliffe S, Novakova I, Vachova M, Zapletalova O, et al. Arandomized, double-blind, placebo-controlled, parallel-group, enriched-design study of nabiximols* (Sativex(®)), as add-on therapy, in subjects with refractory spasticity caused by multiple sclerosis. European Journal of Neurology 2011;18:1122-31. CENTRAL

Rintala 2010 {published data only}

Rintala DH, Fiess RN, Tan G, Holmes SA, Bruel BM. Effect of dronabinol on central neuropathic pain after spinal cord injury: a pilot study. American Journal of Physical Medicine & Rehabilitation 2010;89:840-8. CENTRAL

Turcotte 2015 {published data only}

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

Wade 2003 {published data only}

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

Wade 2004 {published data only}

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. Multiple Sclerosis 2004;10:434-41. CENTRAL

Wallace 2015 {published data only}

Wallace MS, Marcotte TD, Umlauf A, Gouaux B, Atkinson JH. Efficacy of inhaled cannabis on painful diabetic neuropathy. Journal of Pain 2015;16:616-27. CENTRAL

Wilsey 2008 {published data only}

Wilsey B, Marcotte T, Tsodikov A, Millman J, Bentley H, Donaghe H. A randomized, placebo-controlled, crossover trial of cannabis cigarettes in neuropathic pain. Journal of Pain 2008;9:506-21. CENTRAL

Wilsey 2013 {published data only}

Wilsey B, Marcotte T, Deutsch R, Gouaux B, Sakai S, Gouaux B, et al. Low-dose vaporized cannabis significantly improves neuropathic pain. Journal of Pain 2013;14:136-48. CENTRAL

Wissel 2006 {published data only}

Wissel J, Haydn T, Müller J, Brenneis C, Berger T, Berger T. Low dose treatment with the synthetic cannabinoid Nabilone significantly reduces spasticity-related pain: a double-blind placebo-controlled cross-over trial. Journal of Neurology 2006;253:1337-41. CENTRAL

Zajicek 2003 {published data only}

Zajicek J, Fox P, Sanders H, Wright D, Vickery J, Nunn A, et al. Cannabinoids for treatment of spasticity and other symptoms related to multiple sclerosis (CAMS study): multicentre randomized placebo-controlled trial. Lancet 2003;362:1517-26. CENTRAL

Zajicek 2012 {published data only}

Zajicek JP, Hobart JC, Slade A, Barnes D, Mattison PG, MUSEC Research Group. Multiple sclerosis and extract of cannabis: results of the MUSEC trial. Journal of Neurology, Neurosurgery and Psychiatry 2012;83:1125-32. CENTRAL

References to studies awaiting assessment

NCT00699634 {published data only}

NCT00699634. Nabilone for the treatment of phantom limb pain. clinicaltrials.gov/ct2/show/NCT00699634?term=NCT00699634&rank=1 (first posted 18 June 18). CENTRAL

NCT01035281 {published data only}

NCT01035281. Efficacy study of nabilone in the treatment of diabetic peripheral neuropathic pain. clinicaltrials.gov/ct2/show/NCT01035281?term=NCT01035281&rank=1 (first posted 18 December 2009). CENTRAL

NCT01222468 {published data only}

NCT01222468. Effect of cannabinoids on spasticity and neuropathic pain in spinal cord injured persons. clinicaltrials.gov/ct2/show/NCT01222468?term=NCT01222468&rank=1 (first posted 18 October 2010). CENTRAL

Ablin 2016

Ablin J, Ste-Marie PA, Schäfer M, Häuser W, Fitzcharles MA. Medical use of cannabis products: lessons to be learned from Israel and Canada. Schmerz 2016;30:3-13.

AlBalawi 2013

AlBalawi Z, McAlister FA, Thorlund K, Wong M, Wetterslev J. Random error in cardiovascular meta-analyses: how common are false positive and false negative results? Interational Journal of Cardiology 2013;168(2):1102-7. [DOI: 10.1016/j.ijcard.2012.11.048]

Andersohn 2008

Andersohn F, Garbe E. Pharmacoepidemiological research with large health databases. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2008;51:1134-55.

Andreae 2015

Andreae MH, Carter GM, Shaparin N, Suslov K, Ellis RJ, Ware MA, et al. Inhaled cannabis for chronic neuropathic pain: a meta-analysis of individual patient data. Journal of Pain 2015;16(12):1221-32.

Attal 2016

Attal N, de Andrade DC, Adam F, Ranoux D, Teixeira MJ, Galhardoni R, et al. Safety and efficacy of repeated injections of botulinum toxin A in peripheral neuropathic pain (BOTNEP): a randomised, double-blind, placebo-controlled trial. Lancet Neurology 2016;15:555-65.

Baron 2004

Baron R, Binder A. How neuropathic is sciatica? The mixed pain concept [Wie neuropathisch ist die lumboischialgie? Das gemischte schmerz konzept]. Orthopäde 2004;33:568-75.

Baron 2012

Baron R, Wasner G, Binder A. Chronic pain: genes, plasticity, and phenotypes. Lancet Neurology 2012;11(1):19-21. [DOI: 10.1016/S1474-4422(11)70281-]

Baron 2017

Baron R, Maier C, Attal N, Binder A, Bouhassira D, Cruccu G, et al. Peripheral neuropathic pain: a mechanism-related organizing principle based on sensory profiles. Pain 2017;158:261-72.

Berger 2004

Berger A, Dukes EM, Oster G. Clinical characteristics and economic costs of patients with painful neuropathic disorders. Journal of Pain 2004;5(3):143-9. [DOI: 10.1016/j.jpain.2003.12.004]

Berger 2009

Berger A, Toelle T, Sadosky A, Dukes E, Edelsberg J, Oster G. Clinical and economic characteristics of patients with painful neuropathic disorders in Germany. Pain Practice 2009;9(1):8-17. [DOI: 10.1111/j.1533-2500.2008.00244.x]

Berger 2012

Berger A, Sadosky A, Dukes E, Edelsberg J, Oster G. Clinical characteristics and patterns of healthcare utilization in patients with painful neuropathic disorders in UK general practice: a retrospective cohort study. BMC Neurology 2012;12:8.

Bouhassira 2008

Bouhassira D, Lantéri-Minet M, Attal N, Laurent B, Touboul C. Prevalence of chronic pain with neuropathic characteristics in the general population. Pain 2008;136:380-7.

Boychuk 2015

Boychuk DG, Goddard G, Mauro G, Orellana MF. The effectiveness of cannabinoids in the management of chronic nonmalignant neuropathic pain: a systematic review. Journal of Oral and Facial Pain and Headache 2015;29(1):7-14.

Calvo 2012

Calvo M, Dawes JM, Bennett DL. The role of the immune system in the generation of neuropathic pain. Lancet Neurology 2012;11(7):629-42. [DOI: 10.1016/S1474-4422(12)70134-5]

Clauw 2015

Clauw D. What is the meaning of "small fiber neuropathy" in fibromyalgia? Pain 2015;156(11):2115-6.

Cochrane PaPaS 2012

Cochrane Pain, Palliative and Supportive Care Group. PaPaS author and referee guidance. papas.cochrane.org/papas-documents (accessed 5 June 2015).

Cohen 1988

Cohen J. Statistical Power Analysis for the Behavioral Sciences. Hillsdale: Lawrence Erlbaum Associates, 1988.

De Vries 2014

De Vries M, Van Rijckevorsel DC, Wilder-Smith OH, Van Goor H. Dronabinol and chronic pain: importance of mechanistic considerations. Expert Opinion on Pharmacotherapy 2014;15:1525-34.

Dechartes 2013

Dechartres A, Trinquart L, Boutron I, Ravaud P. Influence of trial sample size on treatment effect estimates: meta-epidemiological study. BMJ 2013;346:f2304. [DOI: 10.1136/bmj.f2304]

Dechartres 2014

Dechartres A, Altman DG, Trinquart L, Boutron I, Ravaud P. Association between analytic strategy and estimates of treatment outcomes in meta-analyses. JAMA 2014;312:623-30. [DOI: 10.1001/jama.2014.8166]

Demant 2014

Demant DT, Lund K, Vollert J, Maier C, Segerdahl M, Finnerup NB, et al. The effect of oxcarbazepine in peripheral neuropathic pain depends on pain phenotype: a randomised, double-blind, placebo-controlled phenotype-stratified study. Pain 2014;155(11):2263-73. [DOI: 10.1016/j.pain.2014.08.014]

Derry 2012

Derry S, Moore RA. Topical capsaicin (low concentration) for chronic neuropathic pain in adults. Cochrane Database of Systematic Reviews 2012, Issue 9. Art. No: CD010111. [DOI: 10.1002/14651858.CD010111]

Derry 2014

Derry S, Wiffen PJ, Moore RA, Quinlan J. Topical lidocaine for neuropathic pain in adults. Cochrane Database of Systematic Reviews 2014, Issue 7. Art. No: CD010958. [DOI: 10.1002/14651858.CD010958.pub2]

Derry 2017

Derry S, Rice ASC, Cole P, Tan T, Moore RA. Topical capsaicin (high concentration) for chronic neuropathic pain in adults. Cochrane Database of Systematic Reviews 2017, Issue 1. Art. No: CD007393. [DOI: 10.1002/14651858.CD007393.pub4]

Dworkin 2008

Dworkin RH, Turk DC, Wyrwich KW, Beaton D, Cleeland CS, Farrar JT, et al. Interpreting the clinical importance of treatment outcomes in chronic pain clinical trials: IMMPACT recommendations. Journal of Pain 2008;9(2):105-21. [DOI: 10.1016/j.jpain.2007.09.005]

Dworkin 2013

Dworkin RH, O'Connor AB, Kent J, Mackey SC, Raja SN, Stacey BR, et al. Interventional management of neuropathic pain: NeuPSIG recommendations. Pain 2013;154:2249-61.

Elbourne 2002

Elbourne DR, Altman DG, Higgins JP, Curtin F, Worthington HV, Vail A. Meta-analyses involving cross-over trials: methodological issues. International Journal of Epidemiology 2002;31(1):140-9.

European Medicines Agency 2007

Guideline on clinical medicinal products intended for the treatment of neuropathic pain. www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2009/09/WC500003478.pdf 2007 (Accessed May 2, 2015).

Fayers 2014

Fayers PM, Hays RD. Don't middle your MIDs: regression to the mean shrinks estimates of minimally important differences. Quality of Life Research 2014;23:1-4.

Finnerup 2013

Finnerup NB, Scholz J, Attal N, Baron R, Haanpää M, Hansson P, et al. Neuropathic pain needs systematic classification. European Journal of Pain 2013;17(7):953-6. [DOI: 10.1002/j.1532-2149.2012.00282.x]

Finnerup 2015

Finnerup NB, Attal N, Haroutounian S, McNicol E, Baron R, Dworkin RH, et al. Pharmacotherapy for neuropathic pain in adults: a systematic review and meta-analysis. Lancet Neurology 2015;14(2):162-73. [DOI: 10.1016/S1474-4422(14)70251-0]

Fitzcharles 2014

Fitzcharles MA, Clauw DJ, Ste-Marie PA, Shir Y. The dilemma of medical marijuana use by rheumatology patients. Arthritis Care and Research 2014;66:797-801.

Furukawa 2005

Furukawa TA, Cipriani A, Barbui C, Brambilla P, Watanabe N. Imputing response rates from means and standard deviations in meta-analyses. International Clinical Psychopharmacology 2005;20:49-52.

Gaskell 2016

Gaskell H, Derry S, Stannard C, Moore RA. Oxycodone for neuropathic pain in adults. Cochrane Database of Systematic Reviews 2016, Issue 7. Art. No: CD010692. [DOI: 10.1002/14651858.CD010692.pub3]

GRADEpro GDT 2015 [Computer program]

McMaster University (developed by Evidence Prime)GRADEpro GDT. Hamilton (ON): McMaster University (developed by Evidence Prime), 2015. Available at gradepro.org.

Guindon 2009

Guindon J, Hohmann AG. The endocannabinoid system and pain. CNS & Neurological Disorders Drug Targets 2009;8:403-29.

Gustorff 2008

Gustorff B, Dorner T, Likar R, Grisold W, Lawrence K, Schwarz F, et al. Prevalence of self-reported neuropathic pain and impact on quality of life: a prospective representative survey. Acta Anaesthesiologica Scandinavica 2008;52:132-6.

Guyatt 2013a

Guyatt G, Oxman AD, Sultan S, Brozek J, Glasziou P, Alonso-Coello P, et al. GRADE guidelines: 11. Making an overall rating of confidence in effect estimates for a single outcome and for all outcomes. Journal of Clinical Epidemiology 2013;66:151-7.

Guyatt 2013b

Guyatt GH, Oxman AD, Santesso N, Helfand M, Vist G, Kunz R, et al. GRADE guidelines: 12. Preparing summary of findings tables-binary outcomes. Journal of Clinical Epidemiology 2013;66:158-72.

Hall 2008

Hall GC, Carroll D, McQuay HJ. Primary care incidence and treatment of four neuropathic pain conditions: a descriptive study, 2002-2005. BMC Family Practice 2008;9:26.

Helfert 2015

Helfert SM, Reimer M, Höper J, Baron R. Individualized pharmacological treatment of neuropathic pain. Clinical Pharmacology and Therapeutics 2015;97(2):135-42. [DOI: 10.1002/cpt.19]

Higgins 2003

Higgins JPT, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta-analyses. BMJ 2003;327:557-60.

Higgins 2011a

Higgins JPT, Altman DG, Sterne JAC (editors). Chapter 8: Assessing risk of bias in included studies. In: Higgins JPT, Green S (editors). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Higgins 2011b

Higgins JPT, Deeks JJ, Altman DG (editors). Chapter 16: Special topics in statistics. In: Higgins JPT, Green S (editors), Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Hillard 2012

Hillard CJ, Weinlander KM, Stuhr KL. Contributions of endocannabinoid signaling to psychiatric disorders in humans: genetic and biochemical evidence. Neuroscience 2012;204:207-29.

Hoggart 2015

Hoggart B, Ratcliffe S, Ehler E, Simpson KH, Hovorka J, Lejčko J, et al. A multicentre, open-label, follow-on study to assess the long-term maintenance of effect, tolerance and safety of THC/CBD oromucosal spray in the management of neuropathic pain. Journal of Neurology 2015;262:27-40.

Häuser 2012

Häuser W, Bartram C, Bartram-Wunn E, Tölle T. Adverse events attributable to nocebo in randomized controlled drug trials in fibromyalgia syndrome and painful diabetic peripheral neuropathy: systematic review. Clinical Journal of Pain 2012;28:437-51.

Häuser 2016

Walitt B, Klose P, Fitzcharles MA, Häuser W. Cannabinoids for fibromyalgia. Cochrane Database of Systematic Reviews 2016, Issue 7. Art. No: CD011694. [DOI: 10.1002/14651858.CD011694]

Häuser 2017

Häuser W, Fitzcharles MA, Radbruch L, Petzke F. Efficacy, tolerability and safety of cannabinoids in pain medicine and palliative care: an overview of systematic reviews and of prospective observational studies. Deutsches Ärzteblatt International 2017;114:627-34.

Häuser 2018

Häuser W, Petzke F, Fitzcharles MA. Efficacy, tolerability and safety of cannabis-based medicines for chronic pain management - an overview of systematic reviews. European Journal of Pain2018;22:455-70.

International Council for Harmonisation 2016

International Council for Harmonisation. Medical Dictionary for Regulatory Activities Version 19.1. www.meddra.org/news-and-events/news/all-translations-meddra-version-191-are-now-available2016.

Jensen 2011

Jensen TS, Baron R, Haanpää M, Kalso E, Loeser JD, Rice AS, et al. A new definition of neuropathic pain. Pain2011;152(10):2204-5. [DOI: 10.1016/j.pain.2011.06.017]

Jensen 2015

Jensen B, Chen J, Furnish T, Wallace M. Medical marijuana and chronic pain: a review of basic science and clinical evidence. Current Pain Headache Reports 2015;19:524.

Kalant 2001

Kalant H. Medicinal use of cannabis: history and current status. Pain Research & Management 2001;6:80-91.

Kalso 2013

Kalso E, Aldington DJ, Moore RA. Drugs for neuropathic pain. BMJ 2013;347:f7339.

Katusic 1991

Katusic S, Williams DB, Beard CM, Bergstralh EJ, Kurland LT. Epidemiology and clinical features of idiopathic trigeminal neuralgia and glossopharyngeal neuralgia: similarities and differences, Rochester, Minnesota,1945-1984. Neuroepidemiology 1991;10:276-81.

Koopman 2009

Koopman JS, Dieleman JP, Huygen FJ, De Mos M, Martin CG, Sturkenboom MC. Incidence of facial pain in the general population. Pain 2009;147:122-7.

Koppel 2014

Koppel BS, Brust JC, Fife T, Bronstein J, Youssof S, Gronseth G, et al. Systematic review: efficacy and safety of medical marijuana in selected neurologic disorders: report of the Guideline Development Subcommittee of the American Academy of Neurology. Neurology 2014;82:1556-63.

Krcevski‐Skvarc 2018

Krcevski-Skvarc N, Wells C, Häuser W. Availability and approval of cannabis- based medicines for chronic pain management and palliative/supportive care in Europe – a survey of the status in the chapters of the European Pain Federation. European Journal Pain2018;22:440-54.

L'Abbé 1987

L'Abbé KA, Detsky AS, O'Rourke K. Meta-analysis in clinical research. Annals of Internal Medicine 1987;107:224-33.

Lee 2013

Lee MC, Ploner M, Wiech K, Bingel U, Wanigasekera V, Brooks J, et al. Amygdala activity contributes to the dissociative effect of cannabis on pain perception. Pain 2013;134:123-34.

Long 2009

Long JZ, Nomura DK, Vann RE, Walentiny DM, Booker L, Jin X, et al. Dual blockade of FAAH and MAGL identifies behavioral processes regulated by endocannabinoid crosstalk in vivo. Proceedings of the National Academy of Science of the United States of America 2009;106:20,270-5.

Lunn 2014

Lunn MP, Hughes RA, Wiffen PJ. Duloxetine for treating painful neuropathy, chronic pain or fibromyalgia. Cochrane Database of Systematic Reviews 2014, Issue 1. Art. No: CD007115. [DOI: 10.1002/14651858.CD007115.pub3]

Lynch 2011

Lynch M, Campbell F. Cannabinoids for treatment of chronic non-cancer pain; a systematic review of randomized trials. British Journal of Clinical Pharmacology 2011;72:735-44.

McQuay 1998

McQuay H, Moore R. An Evidence-Based Resource for Pain Relief. Oxford: Oxford University Press, 1998. [ISBN: 0-19-263048-2]

McQuay 2007

McQuay HJ, Smith LA, Moore RA. Chronic pain. In: In: Stevens A, Raftery J, Mant J, Simpson S, editor(s). Health Care Needs Assessment, 3rd Series. Radcliffe Publishing, 2007.

Mintzes 2015

Mintzes B, Lexchin J, Quintano AS. Clinical trial transparency: many gains but access to evidence for new medicines remains imperfect. British Medical Bulletin 2015;116:43-53.

Moher 2009

Moher D, Liberati A, Tetzlaff J, Altman DG, The PRISMA Group (2009). Preferred reporting items for systematic reviews and meta-analyses: The PRISMA Statement. PLoS Medicine 2009;6(7):e1000097. [DOI: 10.1371/journal.pmed1000097]

Moore 1998

Moore RA, Gavaghan D, Tramèr MR, Collins SL, McQuay HJ. Size is everything - large amounts of information are needed to overcome random effects in estimating direction and magnitude of treatment effects. Pain 1998;78(3):209-16. [DOI: 10.1016/S0304-3959(98)00140-7]

Moore 2008

Moore RA, Barden J, Derry S, McQuay HJ. Managing potential publication bias. In: McQuay HJ, Kalso E, Moore RA, editors(s). Systematic Reviews in Pain Research: Methodology Refined. Seattle: IASP Press, 2008:15-24. [ISBN: 978-0-931092-69-5]

Moore 2009

Moore RA, Straube S, Wiffen PJ, Derry S, McQuay HJ. Pregabalin for acute and chronic pain in adults. Cochrane Database of Systematic Reviews 2009, Issue 3. Art. No: CD007076. [DOI: 10.1002/14651858.CD007076.pub2]

Moore 2010a

Moore RA, Eccleston C, Derry S, Wiffen P, Bell RF, Straube S, et al. "Evidence" in chronic pain - establishing best practice in the reporting of systematic reviews. Pain 2010;150(3):386-9. [DOI: 10.1016/j.pain.2010.05.011]

Moore 2010b

Moore RA, Straube S, Paine J, Phillips CJ, Derry S, McQuay HJ. Fibromyalgia: moderate and substantial pain intensity reduction predicts improvement in other outcomes and substantial quality of life gain. Pain 2010;149(2):360-4. [DOI: 10.1016/j.pain.2010.02.039]

Moore 2010c

Moore RA, Moore OA, Derry S, Peloso PM, Gammaitoni AR, Wang H. Responder analysis for pain relief and numbers needed to treat in a meta-analysis of etoricoxib osteoarthritis trials: bridging a gap between clinical trials and clinical practice. Annals of the Rheumatic Diseases 2010;69(2):374-9. [DOI: 10.1136/ard.2009.107805]

Moore 2010d

Moore RA, Smugar SS, Wang H, Peloso PM, Gammaitoni A. Numbers-needed-to-treat analyses - do timing, dropouts, and outcome matter? Pooled analysis of two randomized, placebo-controlled chronic low back pain trials. Pain 2010;151(3):592-7. [DOI: 10.1016/j.pain.2010.07.013]

Moore 2011a

Moore RA, Straube S, Paine J, Derry S, McQuay HJ. Minimum efficacy criteria for comparisons between treatments using individual patient meta-analysis of acute pain trials: examples of etoricoxib, paracetamol, ibuprofen, and ibuprofen/paracetamol combinations after third molar extraction. Pain 2011;152(5):982-9.

Moore 2011b

Moore RA, Mhuircheartaigh RJ, Derry S, McQuay HJ. Mean analgesic consumption is inappropriate for testing analgesic efficacy in post-operative pain: analysis and alternative suggestion. European Journal of Anaesthesiology 2011;28(6):427-32. [DOI: 10.1097/EJA.0b013e328343c569]

Moore 2012

Moore RA, Straube S, Eccleston C, Derry S, Aldington D, Wiffen P, et al. Estimate at your peril: imputation methods for patient withdrawal can bias efficacy outcomes in chronic pain trials using responder analyses. Pain 2012;153(2):265-8. [DOI: 10.1016/j.pain.2011.10.004]

Moore 2013a

Moore RA, Straube S, Aldington D. Pain measures and cut-offs - 'no worse than mild pain' as a simple, universal outcome. Anesthesia 2013;68:400-12.

Moore 2013b

Moore A, Derry S, Eccleston C, Kalso E. Expect analgesic failure; pursue analgesic success. BMJ 2013;346:f2690. [DOI: 10.1136/bmj.f2690]

Moore 2014a

Moore RA, Derry S, Taylor RS, Straube S, Phillips CJ. The costs and consequences of adequately managed chronic non-cancer pain and chronic neuropathic pain. Pain Practice 2014;14(1):79-94. [DOI: 10.1111/papr.12050]

Moore 2014b

Moore RA, Cai N, Skljarevski V, Tölle TR. Duloxetine use in chronic painful conditions - individual patient data responder analysis. European Journal of Pain 2014;18(1):67-75. [DOI: 10.1002/j.1532-2149.2013.00341.x]

Moore 2015a

Moore RA, Chi CC, Wiffen PJ, Derry S, Rice ASC. Oral nonsteroidal anti-inflammatory drugs for neuropathic pain. Cochrane Database of Systematic Reviews 2015, Issue 10. Art. No: CD010902. [DOI: 10.1002/14651858.CD010902.pub2]

Moore 2015b

Moore RA, Derry S, Aldington D, Cole P, Wiffen PJ. Amitriptyline for neuropathic pain in adults. Cochrane Database of Systematic Reviews 2015, Issue 7. Art. No: CD008242. [DOI: 10.1002/14651858.CD008242.pub3]

Moore 2017

Wiffen PJ, Derry S, Bell RF, Rice ASC, Tölle TR, Phillips T, et al. Gabapentin for chronic neuropathic pain in adults. Cochrane Database of Systematic Reviews 2017, Issue 6. Art. No: CD007938. [DOI: 10.1002/14651858.CD007938.pub4]

Moulin 2014

Moulin D, Boulanger A, Clark AJ, Clarke H, Dao T, Finley GA, et al. Pharmacological management of chronic neuropathic pain: revised consensus statement from the Canadian Pain Society. Pain Research & Management 2014;19:328-35.

NICE 2013

National Institute for Health and Care Excellence (NICE). Neuropathic pain - pharmacological management: the pharmacological management of neuropathic pain in adults in non-specialist settings, 2013. www.nice.org.uk/guidance/cg173 (accessed 19 October 2014).

Nüesch 2010

Nüesch E, Trelle S, Reichenbach S, Rutjes AW, Tschannen B, Altman DG, et al. Small study effects in meta-analyses of osteoarthritis trials: meta-epidemiological study. BMJ 2010;341:c3515. [DOI: 10.1136/bmj.c3515]

Owens 2015

Owens B. Drug development: the treasure chest. Nature 2015;525:S6-S8.

Petzke 2016

Petzke F, Enax-Krumova EK, Häuser W. Efficacy, tolerability and safety of cannabinoids for chronic neuropathic pain: a systematic review of randomized controlled studies. Schmerz 2016;30:62-88.

Rappaport 1994

Rappaport ZH, Devor M. Trigeminal neuralgia: the role of self-sustaining discharge in the trigeminal ganglion. Pain 1994;56:127-38.

RevMan 2014 [Computer program]

Nordic Cochrane Centre, The Cochrane CollaborationReview Manager 5 (RevMan 5). Version 5.3. Copenhagen: Nordic Cochrane Centre, The Cochrane Collaboration, 2014.

Schaefert 2015

Schaefert R, Welsch P, Klose P, Sommer C, Petzke F, Häuser W. Opioids in chronic osteoarthritis pain. A systematic review and meta-analysis of efficacy, tolerability and safety in randomized placebo-controlled studies of at least 4 weeks duration [Opioide bei chronischem arthroseschmerz. Systematische übersicht und metaanalyse der wirksamkeit, verträglichkeit und sicherheit in randomisierten, placebokontrollierten studien über mindestens 4 wochen]. Schmerz 2015;29:47-59.

Schünemann 2011

Schünemann HJ, Oxman AD, Vist GE, Higgins JPT, Deeks JJ, Glasziou P, et al. Chapter 12: Interpreting results and drawing conclusions. In: Higgins JPT, Green S (editors), Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from www.handbook.cochrane.org.

Scott 2006

Scott FT, Johnson RW, Leedham-Green M, Davies E, Edmunds WJ, Breuer J. The burden of herpes zoster: a prospective population based study. Vaccine 2006;24(9):1308-14. [DOI: 10.1016/j.vaccine.2005.09.026]

Smith 2016

Smith SM, Amtmann D, Askew RL, Gewandter JS, Hunsinger M, Jensen MP, et al. Pain intensity rating training: results from an exploratory study of the ACTTION PROTECCT system. Pain 2016;157(5):1056-64.

Sommer 2015

Sommer C, Welsch P, Klose P, Schaefert R, Petzke F, Häuser W. Opioids in chronic neuropathic pain. A systematic review and meta-analysis of efficacy, tolerability and safety in randomized placebo-controlled studies of at least 4 weeks duration [Opioide bei chronischem neuropathischem schmerz. Systematische übersicht und metaanalyse der wirksamkeit, verträglichkeit und sicherheit in randomisierten, placebokontrollierten studien über mindestens 4 wochen]. Schmerz 2015;29:35-46.

Stannard 2016

Stannard C, Gaskell H, Derry S, Aldington D, Cole P, Cooper TE, et al. Hydromorphone for neuropathic pain in adults. Cochrane Database of Systematic Reviews 2016, Issue 5. Art. No: CD011604. [DOI: 10.1002/14651858.CD011604.pub2]

Straube 2008

Straube S, Derry S, McQuay HJ, Moore RA. Enriched enrolment: definition and effects of enrichment and dose in trials of pregabalin and gabapentin in neuropathic pain. A systematic review. British Journal of Clinical Pharmacology 2008;66(2):266-75. [DOI: 10.1111/j.1365-2125.2008.03200.]

Straube 2010

Straube S, Derry S, Moore RA, Paine J, McQuay HJ. Pregabalin in fibromyalgia - responder analysis from individual patient data. BMC Musculoskeletal Disorders 2010;11:150. [DOI: 10.1186/1471-2474-11-150]

Sultan 2008

Sultan A, Gaskell H, Derry S, Moore RA. Duloxetine for painful diabetic neuropathy and fibromyalgia pain: systematic review of randomised trials. BMC Neurology 2008;8:29. [DOI: 10.1186/1471-2377-8-]

Thorlund 2011

Thorlund K, Imberger G, Walsh M, Chu R, Gluud C, Wetterslev J, et al. The number of patients and events required to limit the risk of overestimation of intervention effects in meta-analysis--a simulation study. PLOS One 2011;6(10):e25491. [DOI: 10.1371/journal.pone.0025491]

Thyson 2014

Tyson SF, Brown P. How to measure pain in neurological conditions? A systematic review of psychometric properties and clinical utility of measurement tools. Clinical Rehabilitation 2014;28:669-86.

Torrance 2006

Torrance N, Smith BH, Bennett MI, Lee AJ. The epidemiology of chronic pain of predominantly neuropathic origin. Results from a general population survey. Journal of Pain 2006;7:281-9.

Treede 2008

Treede RD, Jensen TS, Campbell JN, Cruccu G, Dostrovsky JO, Griffin JW, et al. Neuropathic pain: redefinition and a grading system for clinical and research purposes. Neurology 2008;70(18):1630-5. [DOI: 10.1212/01.wnl.0000282763.29778.59]

Turner 2013

Turner RM, Bird SM, Higgins JP. The impact of study size on meta-analyses: examination of underpowered studies in Cochrane Reviews. PLOS One 2013;8(3):e59202. [DOI: 10.1371/journal.pone.0059202]

Van Hecke 2014

Van Hecke O, Austin SK, Khan RA, Smith BH, Torrance N. Neuropathic pain in the general population: a systematic review of epidemiological studies. Pain 2014;155:654-62.

Van Hoek 2009

Van Hoek AJ, Gay N, Melegaro A, Opstelten W, Edmunds WJ. Estimating the cost-effectiveness of vaccination against herpes zoster in England and Wales. Vaccine 2009;27(9):1454-67. [DOI: 10.1016/j.vaccine.2008.12.024]

Volkow 2014

Volkow ND, Baler RD, Compton WM, Weiss SR. Adverse health effects of marijuana use. New England Journal of Medicine 2014;370:2219-27.

von Hehn 2012

von Hehn CA, Baron R, Woolf CJ. Deconstructing the neuropathic pain phenotype to reveal neural mechanisms. Neuron 2012;73(4):638-52. [DOI: 10.1016/j.neuron.2012.02.008]

Vos 2012

Vos T, Flaxman AD, Naghavi M, Lozano R, Michaud C, Ezzati M, et al. Years lived with disability (YLDs) for 1160 sequelae of 289 diseases and injuries 1990-2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet 2012;380(9859):2163-96. [DOI: 10.1016/S0140-6736(12)61729-2]

Ware 2015

Ware MA, Wang T, Shapiro S, Collet JP, COMPASS study team. Cannabis for the management of pain: assessment of safety study (COMPASS). Journal of Pain 2015;16:1233-42.

Whiting 2015

Whiting PF, Wolff RF, Deshpande S, Di Nisio M, Duffy S. Cannabinoids for medical use: a systematic review and meta-analysis. JAMA 2015;313:2456-73.

Wiffen 2013

Wiffen PJ, Derry S, Moore RA, Aldington D, Cole P, Rice ASC, et al. Antiepileptic drugs for neuropathic pain and fibromyalgia - an overview of Cochrane reviews. Cochrane Database of Systematic Reviews 2013, Issue 11. Art. No: CD010567. [DOI: 10.1002/14651858.CD010567.pub2]

Zettl 2016

Zettl UK, Rommer P, Hipp P, Patejdl R. Evidence for the efficacy and effectiveness of THC-CBD oromucosal spray in symptom management of patients with spasticity due to multiple sclerosis. Therapeutic Advances in Neurological Disorders 2016;9:9-30.

Zhang 2015

Zhang J, Echeverry S, Lim TK, Lee SH, Shi XQ, Huang H. Can modulating inflammatory response be a good strategy to treat neuropathic pain? Current Pharmaceutical Design 2015;21:831-9.

References to other published versions of this review

Mücke 2016

Mücke M, Phillips T, Radbruch L, Petzke F, Häuser W. Cannabinoids for chronic neuropathic pain. Cochrane Database of Systematic Reviews 2016, Issue 5. Art. No: CD012182. [DOI: 10.1002/14651858.CD012182]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Bermann 2004

Study characteristics

Methods

Disease: plexus root avulsion with ≥ 1 root affected

Study setting: single‐centre (orthopedic clinic), UK; 2001‐2002

Study design: cross‐over

Study duration: 2‐week baseline, 3 cross‐over periods for 10‐14 days, no washout periods

Participants

Inclusion criteria: pain ≥ 4 on 0‐10 scale, no cannabis use for 7 days prior to inclusion,

Exclusion criteria: schizophrenia, other psychotic illness or significant psychiatric illness, other than depression associated with chronic illness; serious cardiovascular disease; significant renal or hepatic impairment; epilepsy or convulsions; significant history of substance abuse; known adverse reaction to cannabis or the product excipients; surgery within 2 months (6 months for nerve repair). Female patients who were pregnant, lactating or at risk of pregnancy were also excluded.

Participants: N = 48, 46 male, 2 female, mean age 39 (23‐63 years). Pain baseline 7.5 (no SD reported) (scale 0‐10). 45.8% had used cannabis medicinally, 60.4 % recreationally.

Interventions

Study medication: oromucosal spray THC only (27 mg/mL), THC/CBD mix (27/25 mg/mL), maximum 48 sprays/d; placebo spray

Rescue medication: none

Allowed co‐therapies: stable analgesic medication over 4 weeks (fentanyl not allowed, amitriptyline max. 75 mg/d, no further details provided)

Outcomes

Participant‐reported pain relief ≥ 50%: reported (NRS 0‐10, average of the last 7 days)

PGIC much or very much improved: not assessed

Withdrawal due to AE: reported

Serious AE attributed to medication: reported

Participant‐reported pain relief ≥ 30%: not reported; calculated by imputation method (NRS 0‐10, average of the last 7 days)

Mean pain intensity: NRS 0‐10, average of the last 7 days; SD calculated from P value

HRQoL: Pain Disability Index 0‐70; SD calculated from P value

Sleep problems: sleep quality 10‐0; SD calculated from P value

Fatigue: not assessed

Psychological distress: General Health Questionnaire12; SD calculated from P value

Withdrawals due to lack of efficacy: reported

Nervous system disorders‐related AE: incompletely reported (not suited for analysis)

Psychiatric disorders‐related AE: incompeletely reported (not suited for analysis)

Any adverse event: open question at each visit; VAS intoxication score for AE

Notes

Funding: GW Pharmaceuticals and the Royal National Orthopaedic Hospital NHS Trust

Conflicts of interest: not declared

"No washout period was used between the three treatment periods. Any carry over effect was unlikely to be for greater than 2–3 days so the first week of titration for each period would be sufficient to counteract any carry over with efficacy comparisons being made by averaging the variables over the last 7 days of treatment".

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Patients were randomly allocated by a computer generated list to the six possible sequences of receiving the three study medications"

Allocation concealment (selection bias)

Low risk

"Although the treatment sequence was blinded, sealed code break envelopes, one for each patient, containing information on the treatment sequence were available if necessary. Blinding was maintained throughout the study".

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

No details reported

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No details reported

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

ITT by LOCF

Selective reporting (reporting bias)

Unclear risk

No study protocol available

Group similarity at baseline

Low risk

Identical demographic and clinical characteristics due to study design

Sample size bias

High risk

< 50 participants per treatment arm

Ellis 2009

Study characteristics

Methods

Disease: HIV neuropathy

Study setting: single‐centre, university, USA; years of study not reported

Study design: cross‐over

Study duration: 2 weeks with 5 treatment days per each period, 2 weeks washout

Participants

Inclusion criteria: adults with documented HIV infection, neuropathic pain refractory to ≥ 2 previous analgesics, and an average score of ≥ 5 on the pain intensity subscale of the Descriptor Differential

Exclusion criteria: (1) current DSM‐IV substance use disorders; (2) lifetime history of dependence on cannabis; (3) previous psychosis with or intolerance to cannabis‐based medicines; (4) concurrent use of approved cannabinoid medications (i.e. Marinol); (5) positive urine toxicology screen for cannabis‐based medicines during the wash‐in week before initiating study treatment; and (6) serious medical conditions that might affect participant safety or the conduct of the trial. Individuals with a previous history of alcohol or other drug dependence were eligible provided that criteria for dependence had not been met within the last 12 months. Participants were excluded if urine toxicology demonstrated ongoing use of non prescribed, recreational drugs such as methamphetamine and cocaine

Treatment group (delta‐9‐THC)/placebo group: N = 34 participants, mean age 49.1 years (SD 6.9); male 100%; pain baseline 11.1 (no SD reported) on a 0‐20 scale; 91% with previous cannabis experience

Interventions

Study medication: smoked cannabis with THC ranging from 4% to 8% provided by the National Institute on Drug Abuse, depending on efficacy and tolerability. Cigarettes without THC. 4 smoking sessions in the 8‐h study day

Rescue medication: not reported

Allowed co‐therapies: stable regimen of opioids, anticonvulsants, antidepressants and analgesics

Outcomes

Participant‐reported pain relief ≥ 50%: not reported and not calculable by imputation method

PGIC much or very much improved: not assessed

Withdrawal due to AE: reported

Serious AE: incompletely reported (not suited for meta‐analysis)

Participant‐reported pain relief ≥ 30%: pain quality and impact descriptor differential scale 0‐20; NNTB reported; number of participants extracted from Andreae 2015

Mean pain intensity: pain quality and impact descriptor differential scale 0‐20; SD calculated from P values

HRQoL: Sickness Impact profile; no details reported (not suited for meta‐analysis)**

Sleep problems: not assessed

Fatigue: not assessed

Psychological distress: BSI**

Withdrawals due to lack of efficacy: not reported

Any adverse event: no details of assessment reported

Nervous system disorders‐related AE: incompletely reported (not suited for meta‐analysis)

Psychiatric disorders‐related AE: incompletely reported (not suited for meta‐analysis)

Notes

Funding: Grant C00‐SD‐104 from the University of California, Center for Medicinal Cannabis Research

Conflicts of interest: Heather Bentley and Ben Gouaux are employees of the Center for Medicinal Cannabis Research at the University of California, San Diego, the study sponsor. Ms Bentley is Project Manager for the CMCR and assisted the investigator with regulatory issues, oversight/monitoring, and preparation of the manuscript. Mr. Gouaux is a Research Associate with the CMCR and assisted the investigator with regulatory issues, oversight/monitoring, data preparation and analysis, and preparation and submission of the article. The study authors declare that over the past 3 years Dr. Atkinson has received compensation from Eli Lilly Pharmaceuticals.

"There was no evident sequence effect"

**No data shown; "As measured by the SIP and BSI, there were similar improvements in total mood disturbance, physical disability and quality of life for the cannabis and placebo treatment"

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Random number generator"

Allocation concealment (selection bias)

Low risk

"Randomization was performed by a research pharmacist ... and the key to study assignment was withheld from investigators until completion statistical analyses".

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

No details reported

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No details reported

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Completer analysis of 30% pain reduction as reported by Andreae 2015

Selective reporting (reporting bias)

High risk

Some outcomes were not reported

Group similarity at baseline

Low risk

Identical clinical and demographic characteristics due to study design

Sample size bias

High risk

< 50 participants per treatment arm

Frank 2008

Study characteristics

Methods

Disease: chronic central and PNP (radiculopathy, CRPS, diabetic neuropathy, posttraumatic or postsurgery, trigeminal neuralgia, PHN)

Study setting: outpatient units of 3 hospitals in the UK, 2001‐2002

Study design: cross‐over

Study duration: 1 week pre study, 6 weeks treatment, washout 2 weeks, 6 weeks treatment

Participants

Inclusion criteria: pain, such as burning, stabbing, or paraesthesia within the distribution of a peripheral nerve and a clear clinical history of its cause (sensory abnormality, allodynia, burning pain, lancinating pain, sympathetic dysfunction), pain ≥ 40 on a 100 mm VAS, stable medication

Exclusion criteria: DHC not stopped 2 weeks prior to inclusion, antipsychotics, benzodiazepines (except for night sedation), MAO inhibitors, legal action, ongoing cannabis‐based medicines, severe hepatic or renal disease, epilepsy, bipolar disorder, psychosis, or a history of substance misuse

Participants: DHC then nabilone: N = 48 participants, mean age 50.6 (SD 15.2) years. 23 female. Mean pain baseline 69.6 (range 29‐95) on a 0‐100 scale. No reports on prior use of cannabis.

Participants: nabilone then DHC: N = 48, mean age 49.7 (SD 12.0), 27 male, 21 female; Mean pain baseline 69.6 (range 29‐95) on a 0‐100 scale. No reports on prior use of cannabis.

Interventions

Study medication: dose adjustment every week (twice first week) from 30‐240 mg DHC and 0.25‐2 mg nabilone

Rescue medication: paracetamol 500 mg and codeine 30 mg throughout washout up to 8 times/d

Allowed co‐therapies: "Stable analgesics"

Outcomes

Participant‐reported pain relief ≥ 50%: not reported, calculated by imputation method (daily pain score summarised as last bi‐weekly means VAS 0‐100)

PGIC much or very much improved: not assessed

Withdrawal due to AE: reported

Serious AE attributed to medication: reported

Participant‐reported pain relief ≥ 30%: not reported, calculated by imputation method (daily pain score summarised as last bi‐weekly means VAS 0‐100)

HRQoL: SF‐36 physical functioning 50‐0

Sleep problems: NRS 0‐10; data reported not suited for meta‐analysis (P = 0.20)

Fatigue: not assessed

Psychological distress: SF‐36 Mental Health 50‐0; data reported not suited for meta‐analysis (P = 0.20)

Withdrawals due to lack of efficacy: not reported

Withdrawal due to AE: reported

Any adverse event: "At each visit the patients filled in a side effects assessment form"

Nervous system disorders‐related AE: incompletely reported, not suited for meta‐analysis

Psychiatric disorders‐related AE:incompletely reported, not suited for meta‐analysis

Notes

Funding: grant from Cambridge Laboratories

Conflict of Interest: BF’s salary was provided as part of the above research grant although he was employed by the Newcastle upon Tyne University Hospitals Trust.

"We excluded carry over by basing the analyses from the last two weeks of each treatment period".

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Treatment was allocated by random permuted blocks of 10, stratified by centre."

Allocation concealment (selection bias)

Low risk

"The pharmacies at the treatment centres, the patients, and all clinical personnel involved in the trial were unaware of treatment allocation at all times." Code breaking envelopes

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"The pharmacy at St Mary’s Hospital supplied identical white capsules containing 250 μg nabilone or 30 mg dihydrocodeine."

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No information provided

Incomplete outcome data (attrition bias)
All outcomes

High risk

Available cases analysis (all participants randomised, which provided data in each treatment period)

Selective reporting (reporting bias)

Low risk

All outcomes reported as outlined in the study protocol ISRCTN15330757

Group similarity at baseline

Low risk

Similar demographic and clinical characteristics at baseline

Sample size bias

Unclear risk

50‐199 participants per treatment arm

Langford 2013

Study characteristics

Methods

Disease: central neuropathic pain in multiple sclerosis (MS)

Study setting: multicentre, 33 sites in UK, Canada, Spain, France, Czech Republic; 2006‐2008

Study design: Patients who had failed to gain adequate analgesia from existing medication were treated with THC/CBD spray or placebo as an add‐on treatment in a double‐blind manner, for 14 weeks to investigate the efficacy of the medication in MS‐induced neuropathic pain. This parallel‐group phase of the study was then followed by an 18‐week randomised withdrawal study (14‐week, open‐label treatment period plus a double‐blind, 4‐week, randomised‐withdrawal phase)

Study duration: Phase A: 1‐week baseline, 14‐week treatment. Phase B: 14‐week, open treatment phase with 2 weeks' titration and 12 weeks' stable dose, followed by a randomised withdrawal phase of four weeks (only in France and Czech Republic)

Participants

Inclusion criteria: chronic neuropathic pain due to MS, of at least 3 months' duration. Participants were also to have a sum score of at least 24 on a pain 0–10 point NRS on the last 6 days during the baseline period. In addition, their analgesic regimen was to be stable for at least 2 weeks preceding the study entry day. For Phase B also: ≥ 3 sprays/d in last 7 days of phase A, and tolerability (that means no AE), stable medication

Exclusion criteria: other somatic pain causes with severe pain, including PNP, significant psychiatric (except depression related to pain), renal, hepatic, cardiovascular, or convulsive disorders, sensitivity to cannabis‐based medicines

Phase A, treatment group: N = 167, female/male (54/113), mean age 48.42 (SD 10.43), 11 (7%) with cannabis experience

Placebo group: N = 172, male/female (55/117), mean age 49.51 (SD 10.50) 10 (6%) with cannabis experience

Phase B, treatment group: N = 21; female/male (11/10), mean age 46.2 (10.39), 0 patients with cannabis experience

Placebo group: N = 21, female/male 14/7, mean age 49.82 (9.75), 1 patient with cannabis experience

Interventions

Study medication: THC/CBD oromucosal spray. Each actuation of active medication delivered 2.7 mg of THC and 2.5 mg of CBD to the oral mucosa. Placebo delivered the excipient plus colorants. Max. 12 sprays/24 h

Rescue medication: paracetamol

Allowed co‐therapies: pain medication: stable for at least 2 weeks

Outcomes

Participant‐reported pain relief ≥ 50% (parallel): only OR reported, calculated by imputation method (NRS 0‐10 for mean daily chronic neuropathic pain, average over 7 days at baseline and final 7 days)

PGIC much or very much improved (parallel): reported

Withdrawal due to AE (parallel): reported

Serious AE (parallel and EERW): reported

Participant‐reported pain relief ≥ 30%:: reported

Mean pain intensity (parallel): NRS 0‐10 for mean daily chronic neuropathic pain, average over 7 days at baseline and final 7 days; SD calculated from P value

HRQoL (parallel): EQ‐5D VAS 0‐100

Sleep problems (parallel): NRS 0‐10; SD calculated from P value

Fatigue: NRS 0‐10; SD calculated from P value

Psychological distress (parallel): SF‐36 mental health: SD calculated from P value

Withdrawals due to lack of efficacy (parallel): reported

Any adverse event (parallel and EERW): reported. Details of assessment of AEs not reported.

Nervous system disorders‐related AE (parallel and EERW): reported

Psychiatric disorders‐related AE (parallel and EERW): reported

Notes

Funding: GW Pharmaceuticals

Conflicts of interest: R. Langford, J. Mares, A. Novotna, M. Vachora, I. Novakova, W. Notcutt, and S. Ratcliffe were all investigators in this study and their organizations received investigator fees from GW Pharma Ltd. accordingly for their participation in the study. R. Langford, W. Notcutt, and S. Ratcliffe have received consultancy and speaker fees from GW Pharma Ltd. to attend meetings.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Randomization occurred using a pre‐determined computer generated randomisation code in which treatment allocation was stratified by centre, and used randomly permuted blocks of variable sizes. Separate randomisation schemes, using the same strategy, were produced for each part of the study."

Allocation concealment (selection bias)

Low risk

Separate randomisation schemes

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

No details reported

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No details reported

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

ITT by LOCF

Selective reporting (reporting bias)

Low risk

Data reported as outlined in the study protocol NCT00391079 available

Group similarity at baseline

Low risk

Similar demographic and clinical characteristics at baseline

Sample size bias

Unclear risk

50‐199 participants per treatment arm

Lynch 2014

Study characteristics

Methods

Disease: chemotherapy‐induced neuropathic pain

Study setting: single centre, Canada; year of study not reported

Study design: cross‐over design

Study duration: 4 weeks each and 2 weeks washout

Participants

Inclusion criteria: neuropathic pain persisting for 3 months after completing chemotherapy with paclitaxel, vincristine, or cisplatin. The average 7‐day intensity of pain had to be ≥ 4 on an 11‐point NRS. Participants also exhibited sensory abnormalities comprising allodynia, hyperalgesia, or hypethesia. Concurrent analgesics had to be stable for 14 days before entry into the trial.

Exclusion criteria: ischaemic heart disease, ongoing epilepsy, a personal or family history of schizophrenia, or psychotic disorder or substance abuse or dependency within the previous 2 years. Exclusion criteria also included pregnancy or other medical condition that might compromise safety in the trial.

Both groups: N = 18; mean age 58 (SD 11.34) years; 15/18 female; previous cannabis use 5/18

Interventions

Study medication: THC/CBD oromucosal spray. Each actuation of active medication delivered 2.7 mg of THC and 2.5 mg of CBD to the oral mucosa. Placebo delivered the excipient plus colorants. Max. 12 sprays/24 h

Rescue medication: not reported

Allowed co‐therapies: pain medication (anticonvulsants, antidepressants, NSAIDs, opioids): stable for at least 2 weeks

Outcomes

Participant‐reported pain relief ≥ 50%: not reported, calculated by imputation method. NRS (0‐10) for mean daily chronic neuropathic pain, average over 7 days at baseline and final 7 days

PGIC much or very much improved: not assessed

Withdrawal due to AE: reported

Serious AE: reported

Participant‐reported pain relief ≥ 30%: not reported, calculated by imputation method

Mean pain intensity: reported

HRQoL (parallel): SF‐36 physical component summary score 50‐0

Sleep problems: not assessed

Fatigue: not assessed

Psychological distress: SF‐36 mental health summary score 50‐0

Withdrawals due to lack of efficacy: not reported

Any adverse event: not reported. No details of assessment of AEs reported.

Nervous system disorders‐related AE: reported (summarised by the authors of the review)

Psychiatric disorders‐related AE: reported (summarised by the authors of the review)

Notes

Funding: none

Conflicts of interest: the study authors declare no conflicts of interest

"Thus, the two week washout was chosen to assure no carry over effect between study arms"

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated randomisation schedule

Allocation concealment (selection bias)

Low risk

"Participants and study staff were blinded to the randomisation code, which was not broken until the completion of the study."

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

No details reported

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No details reported

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

ITT by LOCF

Selective reporting (reporting bias)

Unclear risk

No protocol reported

Group similarity at baseline

Low risk

Identical baseline characteristics due to study design

Sample size bias

High risk

< 50 participants per treatment arm

NCT00710424

Study characteristics

Methods

Disease: painful diabetic neuropathy

Study setting: multicentre international trial, UK, Czech Republic, Romania; July 2005‐2006

Study design: parallel

Study duration: 1 week baseline, 14 weeks treatment

Participants

Inclusion criteria: ciagnosed with Type 1 or 2 diabetes mellitus as diagnosed according to the WHO criteria. Diagnosed with neuropathic pain due to distal symmetrical diabetic neuropathy of at least 6 months' duration, as defined by a NDS score of ≥ 4, and in whom pain was not wholly relieved with their current therapy. The NDS score must be attained from ≥ 2 different test parameters and not only the ankle jerk reflex. The last 6 daily diary 0‐10 NRS pain scores before randomisation summed to at least 24. Stable dose of regular pain medication and non‐pharmacological therapies (including TENS) for ≥ 14 days prior to the screening visit and willingness for these to be maintained throughout the study.

Exclusion criteria: uncontrolled diabetes with HbA1c blood levels of > 11% at Visit 1, Day B1. Had used cannabinoid‐based medications within 60 days of study entry and were unwilling to abstain for the duration for the study. History of schizophrenia, other psychotic illness, severe personality disorder or other significant psychiatric disorder other than depression associated with their underlying condition, known or suspected history of alcohol or substance abuse. History of epilepsy or recurrent seizure, postural drop of 20 mmHg or more in systolic blood pressure at screening. Evidence of cardiomyopathy, MI, cardiac disease. QT interval; of > 450 ms (men) or > 470 ms (women) at Visit 1. Secondary or tertiary atrioventricular block or sinus bradycardia (HR < 50 bpm) or sinus tachycardia (HR > 110 bpm) at Visit 1. Diastolic blood pressure of < 50 mmHg or >105 mmHg in a sitting position at rest for 5 min prior to randomisation. Impaired renal hepatic function

Treatment group: N = 149: mean age 60.8 (10.38 SD) years; female/male 56/93. No reports on baseline pain scores and on previous cannabis use.

Placebo group: N = 148; mean age 58.2 (10.57 SD) years; female/male 58/90. No reports on baseline pain scores and on previous cannabis use.

Interventions

Study medication: Sativex (DHC 27 mg/mL/CBD25 mg/mL), delivered in 100 µL actuations by mucosal spray, maximum max per 24 h: 65 mg TC/60 mg cannabidiol); placebo

Rescue medication: no information provided

Allowed co‐therapies: no information provided

Outcomes

Participant‐reported pain relief ≥ 50%: not reported and not calculable by imputation method. Mean Diabetic Neuropathy Pain 0‐10 NRS score at the end of treatment (average of last 7 days' treatment) (Your nerve pain over the last 24 h from 0‐10);

PGIC much or very much improved: reported

Withdrawal due to AE: reported

Serious AE: reported

Participant‐reported pain relief ≥ 30%:reported

Mean pain intensity: Mean Diabetic Neuropathy Pain 0‐10 NRS score at the end of treatment (average of last 7 days' treatment)

HRQoL:: EQ‐5D 0 ‐100

Sleep problems: NRS 0‐10

Fatigue: not assessed

Psychological distress: not assessed

Withdrawals due to lack of efficacy: reported

Any adverse event: mean intoxication score. No details of assessment reported

Nervous system disorders‐related AE: reported

Psychiatric disorders‐related AE: reported

Notes

Funding: GW Pharmaceuticals

Conflicts of interest: not declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No information provided

Allocation concealment (selection bias)

Unclear risk

No information provided

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Identical placebo

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No information provided.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

ITT by LOCF

Selective reporting (reporting bias)

Low risk

All predefined outcomes reported

Group similarity at baseline

Low risk

Similar demographic characteristics at baseline

Sample size bias

Unclear risk

50‐199 participants per treatment arm

NCT01606176

Study characteristics

Methods

Disease: MS and other defects of neurological function

Study setting: multicentre trial in the UK, no year of study reported

Study design: parallel

Study duration: baseline period, 3‐week treatment period

Participants

Inclusion criteria: chronic refractory pain due to multiple sclerosis or other defects of neurological function. Neuropathic pain with a mean severity NRS score at ≥ 4 during last 7 days of the baseline period. Relatively stable neurology during the preceding 6 months. Stable medication regimen during the preceding 4 weeks. Had not used cannabis‐based medicines for at least the preceding 7 days and willing to abstain from any use of cannabis‐based medicines during the study

Exclusion criteria: history of schizophrenia, other psychotic illness, severe personality disorder or other significant psychiatric disorder other than depression associated with their underlying condition. History of alcohol or substance abuse. Severe cardiovascular disorder, such as ischaemic heart disease, arrhythmias (other than well‐controlled atrial fibrillation), poorly controlled hypertension or severe heart failure. History of autonomic dysreflexia. History of epilepsy. Renal and liver problems

Treatment group (delta‐9‐THC): N = 36, female/male 20/16, mean age 51.72 (SD 12.11), 24 in MS‐subset. No baseline pain scores reported. No reports on previous cannabis use

Placebo group: N = 34, female/male 21/13, mean age 57.61 (SD 10.28), 19 in MS‐subset. No baseline pain scores reported. No reports on previous cannabis use

Interventions

Study medication: each actuation of oromucosal spray delivers 2.5 mg THC and 2.5 mg CBD. The maximum permitted dose of was 8 actuations in any 3‐hour period, and 48 actuations in any 24‐h period (THC 120 mg:CBD 120 mg). Placebo same number of actuations possible

Rescue medication: no details provided, but percentage of days with uses recorded as secondary outcome (less in active group)

Allowed co‐therapies: no details provided

Outcomes

Participant‐reported pain relief ≥ 50%: not reported and not calculable by imputation method. NRS 0‐10, 3 measures/day, average of the last 7 days

PGIC much or very much improved: reported

Withdrawal due to AE: reported, systematic assessment

Serious AE: reported, systematic assessment

HRQoL: Spitzer Quality of life index 15‐0

Participant‐reported pain relief ≥ 30%: not reported and not calculable by imputation method

Mean pain intensity: NRS 0‐10, 3 measures/day, average of the last 7 days

Sleep problems: NRS 0‐10

Fatigue: not assessed

Psychological distress: not assessed

Withdrawals due to lack of efficacy: not reported

Any adverse event: reported; systematic assessment, no details reported

Nervous system disorders‐related AE: reported; systematic assessment

Psychiatric disorders‐related AE: reported; systematic assessment

Notes

Funding: GW Pharmaceuticals

Conflicts of interest: not declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No information provided

Allocation concealment (selection bias)

Unclear risk

No information provided

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"Identical placebo"

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No information provided

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

ITT by LOCF

Selective reporting (reporting bias)

Low risk

All predefined outcomes reported

Group similarity at baseline

Low risk

Similar demographic characteristics at baseline

Sample size bias

High risk

< 50 participants per treatment arm

NCT01606202

Study characteristics

Methods

Disease: intractable neuropathic pain associated with spinal cord Injury

Study setting: multicentre study UK, Romania; no years of study reported

Study design: parallel

Study duration: 7‐21 days baseline period, 3 weeks treatment

Participants

Inclusion criteria: diagnosis of non‐acute spinal cord injury, with central neuropathic pain not wholly relieved by current therapy. Central neuropathic pain with a mean severity NRS score ≥ 4 during last 7 days of the baseline period. Relatively stable neurology during the preceding 6 months. Stable medication regimen during the preceding 4 weeks. Had not used cannabis‐based medicines for at least the preceding 7 days and willing to abstain from any use of cannabis‐based medicines during the study

Exclusion criteria: history of schizophrenia, other psychotic illness, severe personality disorder or other significant psychiatric disorder other than depression associated with their underlying condition. History of alcohol or substance abuse. Severe cardiovascular disorder, such as ischaemic heart disease, arrhythmias (other than well‐controlled atrial fibrillation), poorly controlled hypertension or severe heart failure. History of autonomic dysreflexia. History of epilepsy. Renal and liver problems

Treatment group (delta‐9‐THC): N = 56, age 48.7 (12.97), female/male 13/43. No reports on pain baseline scores and on previous cannabis use

Placebo group: N = 60, age 47.6 (12.69), female/male 12/48. No reports on pain baseline scores and on previous cannabis use

Interventions

Study medication: THC (27 mg/mL): CBD (25 mg/mL) as extract of Cannabis sativa L., with peppermint oil, 0.05%, in ethanol:propylene glycol (50:50) excipient. Each actuation delivered 100 μL (THC 2.7 mg and CBD 2.5 mg). The maximum permitted dose of study medication was 8 actuations in any 3‐h period, and 48 actuations in any 24‐h period

Rescue medication: paracetamol 500 mg

Allowed co‐therapies: stable medication regimen

Outcomes

Participant‐reported pain relief ≥ 50%: not reported and not calculable by imputation method. NRS 0‐10 Neuropathic Pain Scale

PGIC much or very much improved: reported

Withdrawal due to AE: reported

Serious AE:

Participant‐reported pain relief ≥ 30%:: not reported and not calculable by imputation method

Mean pain intensity: NRS 0‐10 Neuropathic Pain Scale

HRQoL: Spitzer Quality of Life Index Score 15‐0

Sleep problems: sleep disturbance NRS 0‐10

Fatigue: not assessed

Psychological distress: not assessed

Withdrawals due to lack of efficacy: not reported

Any adverse event: reported. No details of assessment reported

Nervous system disorders‐related AE: reported

Psychiatric disorders‐related AE: reported

Notes

Funding: GW Pharmaceuticals

Conflicts of interest: not declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No information provided

Allocation concealment (selection bias)

Unclear risk

No information provided, but identical placebo

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"Identical placebo"

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No information provided

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

ITT by LOCF

Selective reporting (reporting bias)

Low risk

All predefined outcomes reported

Group similarity at baseline

Low risk

Similar demographic characteristics of the study groups at baseline

Sample size bias

Unclear risk

50‐199 participants per treatment arm

Nurmikko 2007

Study characteristics

Methods

Disease: pain and allodynia patients with unilateral neuropathic pain of peripheral origin of various aetiologies

Study setting: multicentre (5 UK, 1 Belgium); study period not reported

Study design: parallel

Study duration: baseline 7‐10 days, therapy 5 weeks

Participants

Inclusion criteria: unilateral PNP and allodynia, at least 6 months with identifiable nerve lesion, unilateral PNP and allodynia, CRPS type II, ≥ 4 on a NRS for spontaneous pain 4 out of 7 days during baseline. A stable medication regimen of analgesics for at least 2 weeks prior to study entry.

Exclusion criteria: cannabinoid use < 7 days, failure to abstain, schizophrenia, psychosis, or other major psychiatric condition beyond depression with underlying condition. Concomitant severe non‐neuropathic pain or the presence of cancer‐related neuropathic pain or from diabetes mellitus, known history of alcohol or substance abuse, severe cardiovascular condition, poorly controlled hypertension, epilepsy, pregnancy, lactation, significant hepatic or renal impairment

Treatment group (delta‐9‐THC): N = 63, female 35, mean age 52.4 (SD 15.8) years. Pain baseline 7.3 (SD 1.4) on 0‐10 scale. 13 (21%) prior cannabis use
Placebo group: N = 62, female 39, age 54.3 (15.2) years; pain baseline 7.2 (SD 1.5) on 0‐10 scale. 2 (19%) prior cannabis use

Interventions

Study medication: spray for sublingual and oro‐pharyngeal administration. Each 100 μL spray delivers 2.7 mg of THC and 2.5 mg of CBD, identically appearing placebo spray. Participants were allowed a maximum dose of 8 sprays per 3‐h interval and a maximum of 48 sprays per 24 h.

Rescue medication: none

Allowed co‐therapies: stable dose regimen

Outcomes

Participant‐reported pain relief ≥ 50%: reported. NRS 0‐10 over 7 days

PGIC much or very much improved: only average scores reported (not suited for meta‐analysis)

Withdrawal due to AE: assessed

Serious AE: assessed; only psychiatric serious AEs reported

Participant‐reported pain relief ≥ 30%: reported. NRS 0‐10 over 7 days

Mean pain intensity: neuropathic pain scale total score 0‐60

HRQoL: not assessed

Sleep problems: NRS 0‐10; SD calculated from P value

Fatigue: not assessed

Psychological distress: General Health Questionnaire 0‐48: SD calculated from P value

Any adverse event: not reported (details of assessment of AE not reported)

Nervous system disorders‐related AE: incompeletely reported (not suited for analysis)

Psychiatric disorders‐related AE: incompeletely reported (not suited for analysis)

Notes

Funding: GW Pharmaceuticals

Conflicts of interest: not declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"After eligibility was confirmed, patients were assigned to the next sequential randomisation number within each centre. The randomisation schedule had a 1:1 treatment allocation ratio with randomly permuted blocks stratified by centre and was generated using a computer based pseudo‐random number algorithm".

Allocation concealment (selection bias)

Low risk

"The randomisation schedule was held by the sponsor with a copy in patient‐specific sealed envelopes sent to the pharmacy in each centre."

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"That the smell and taste of the cannabinoid preparation might lead to unblinding was averted by disguising them with addition of peppermint oil to both preparations."

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"The analyses were verified by an independent statistician. The principal investigator had full access to all the data and carried out further confirmatory analyses"

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

ITT by LOCF

Selective reporting (reporting bias)

Low risk

All predefined outcomes reported

Group similarity at baseline

Low risk

Similar clinical and demographic characteristics at baseline

Sample size bias

Unclear risk

50‐199 participants per treatment arm

Rog 2005

Study characteristics

Methods

Disease: central pain in MS

Study setting: UK, single‐centre; study period not reported

Study design: parallel, randomised, placebo‐controlled, parallel‐group study

Study duration: 5 weeks, including 1 week baseline

Participants

Inclusion criteria: at least 6 months after MS diagnosis, at least 3 months central pain with unlikely other cause, both with dysaesthetic characteristics or painful spasm, 2 weeks of stable analgesic regimen, no cannabinoid use the last 7 days

Exclusion criteria: spasticity‐related pain, visceral pain, headache, acute MS‐related pain, major psychiatric disorder, other than pain‐related depression, severe concomitant illness, seizures, history or suspicion of substance abuse, diabetes mellitus, levodopa use, hypersensitivity to cannabis‐based medicines

Treatment group (delta‐9‐THC/CBD): N = 34; 6 male/28 female, mean age 50.3 (SD 6.7) years; 15 with previous cannabis exposure

Placebo group: N = 32; 8 male/24 female; mean age 48.1 (SD 9.7) years; 21 with previous cannabis exposure

Interventions

Study medication: Oromucosal spray containing 2.7 mg THC and 2.5 mg CBD per 100 µL spray, max 48 sprays in 48 h, identically appearing placebo

Rescue medication: not reported

Allowed co‐therapies: amitriptylin maximally 75 mg/d

Outcomes

Participant‐reported pain relief ≥ 50%: not reported, calculated by imputation method. NRS 0‐10 for most troublesome neuropathic pain at daily maximum, mean of 7 days

PGIC much or very much improved: reported

Withdrawal due to AE: reported

Serious AE attributed to medication: reported

Participant‐reported pain relief ≥ 30%: not reported, calculated by imputation method

Mean pain intensity: NRS 0‐10 for most troublesome neuropathic pain at daily maximum, mean of 7 days

HRQoL: not assessed

Sleep problems: sleep quality 10‐0; SD calculated from P value

Fatigue: not assessed

Psychological distress: General Health Questionnaire 0‐48: SD calculated from P value

Withdrawals due to lack of efficacy: not reported

Any adverse event: not reported. No details of assessment reported

Nervous system disorders‐related AE: reported

Psychiatric disorders‐related AE: reported

Notes

Funding: GW Pharmaceuticals

Conflicts of interest: Rog, Young and Nurmikko received funding and/or honoraria from GW pharmaceuticals

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Pre‐determined randomisation code that remained unknown to study personnel throughout the trial. Randomised permuted blocks of 4

Allocation concealment (selection bias)

Low risk

Pharmacist dispensed medication

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Identically appearing placebo also for smell

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Secondary outcomes assessed by blinded nurses

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

ITT by LOCF

Selective reporting (reporting bias)

Low risk

Consistent reporting of all outcomes

Group similarity at baseline

Low risk

Similar demographic and clinical characteristics at baseline

Sample size bias

High risk

< 50 participants per study arm

Schimrigk 2017

Study characteristics

Methods

Disease: central neuropathic pain in MS

Study setting: single‐centre (Neurology Department), Germany, study period 2007‐2010

Study design: parallel

Study duration: dose titration of study medication over 2 weeks, 2 weeks' titration, followed by a 10‐week maintenance phase. 32 weeks open label

Participants

Inclusion criteria: aged 18–70 years, met the McDonald criteria for definite MS and had stable disease symptoms and moderate‐severe central neuropathic pain (CNP) at maximal pain area for at least 3 months as reported by participants (Numerical Rating Scale (NRS) for pain ≥ 4). CNP was defined as initiated or caused by a primary lesion or dysfunction of the CNS.

Exclusion criteria: any peripheral pain syndromes, pre‐existing psychotic disorders, severe cardiac diseases, or known substance abuse; dronabinol intake within the last 12 months prior to study entry or Marijuana use within 1 month prior to study entry

Treatment group (dronabinol): N = 124, mean age 48.4 (SD 9.6) years, 88% female, time since CNP diagnosis 130 (96) months, pain score baseline (extracted from figure 6.6), previous cannabis use not reported

Placebo group: N = 116, mean age 47.0 (SD 9.7) years, 87% female, time since CNP diagnosis 138 (98) months, pain score baseline (extracted from figure 6.8), previous cannabis use not reported

Interventions

Study medication: dosing was increased every 5 days by 2.5 mg to reach a daily dose between 7.5 and 15.0 mg

Rescue medication: oral intake of tramadol

Allowed co‐therapies: amitriptyline and gabapentin, if started at least 3 months earlier with a stable dose

Outcomes

Participant‐reported pain relief ≥ 50%:: not reported. NRS 0‐10 mean weekly pain score. Calculated by imputation method. Baseline pain scores extracted from figure

PGIC much or very much improved: not assessed

Withdrawal due to AE: reported

Serious AE attributed to medication: reported

Participant‐reported pain relief ≥ 30%:: not reported. NRS 0‐10 mean weekly pain score. Calculated by imputation method. Baseline pain scores extracted from figure

Mean pain intensity: NRS 0‐10 mean weakly pain score

HRQoL: Short form health survey SF‐36. Mean changes without SD or P value reported*

Sleep problems: not assessed

Fatigue: not assessed

Psychological distress: not assessed

Withdrawals due to lack of efficacy: reported

Any adverse event: for safety analysis, vital signs, laboratory parameters, (serious) AEs (SAEs) including (serious) adverse reactions (SARs) were regularly assessed during all 3 periods. Furthermore, participantss rated the global tolerability on a 4‐point rating scale (1 = very good to 4 = poor). If study medication intake was interrupted, the investigator documented withdrawal symptoms such as restlessness, irritability, sleep interference, decreased appetite, excessive sweating, or other drug‐dependence‐related symptoms

Nervous system disorders‐related AE: not reported

Psychiatric disorders‐related AE: not reported

Notes

Funding: Bionorica research GmbH (Innsbruck, Austria)

Conflicts of interest: CN, EMK, GW, and DA‐S are employees of Bionorica SE, Germany. SS has received grant support and speaker honoraria from Bayer Vital, Bionorica, Biogen, BMS, DIAMED, Genzyme, Novartis, Pfizer, Teva. MM has received lecture fees, travel grants and honoraria for consulting from Bayer Health Care AG, Biogen GmbH, Bionorica, Merck Serono, Novartis Pharma GmbH, Sanofi‐Aventis (Genzyme), and Teva

*no significant difference

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated randomization code

Allocation concealment (selection bias)

Unclear risk

No details reported

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

No details reported

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No details reported

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No details reported ("Full analysis set")

Selective reporting (reporting bias)

Low risk

All outcomes as outlined in protocol NCT00959218 reported

Group similarity at baseline

Low risk

Similar demographic and clinical characteristics at baseline

Sample size bias

Unclear risk

50‐199 participants per treatment arm

Selvarajah 2010

Study characteristics

Methods

Disease: Chronic painful diabetic peripheral polyneuropathy in diabetes mellitus type 1 and 2

Study setting: Single‐centre (Diabetes Research Department), UK; study period not reported

Study design: Parallel

Study duration: Dose titration of study medication over 2 weeks, followed by a 10‐week maintenance phase

Participants

Inclusion criteria: Neuropathy Total Symptom Score 6 > 4 and < 16 for at least 6 months with stable glycaemic control (A1C 11%), persistent pain, despite an adequate trial of tricyclic antidepressants

Exclusion criteria: Not reported

Treatment group (delta‐9‐THC/CBD): N = 15, Mean age 58.2 (SD 8.8) years, 4 female, mean diabetes duration 11.2 ± 8.4 years, 2 with previous cannabis use

Placebo group: N = 15, 7 female, mean age 54.4 (SD 11.6) years, mean diabetes duration 13.7 (SD 6) years; 2 with previous cannabis use

Interventions

Study medication: Sativex (tetrahydrocannabinol (27 mg/mL) and CBD (25 mg/mL)) as a pump‐action spray, sublingually, up to 4 doses per day

Rescue medication: Not reported

Allowed co‐therapies: Not reported

Outcomes

Participant‐reported pain relief ≥ 50%: Reported. VAS 0‐10

PGIC much or very much improved: Not assessed

Withdrawal due to AE: Reported, but not the proportion of patients in each group

Serious AE attributed to medication: Not reported

Participant‐reported pain relief ≥ 30%:: Not reported, calculated by imputation method (VAS 0‐10)

Mean pain intensity: Neuropathic pain scale (VAS 0‐100)

HRQoL: EQ‐5D health status index

Sleep problems: Sleep quality 10‐0; SD calculated from P value

Fatigue: Not assessed

Psychological distress: General Health Questionnaire 0‐48: SD calculated from P value

Withdrawals due to lack of efficacy: Not reported

Any adverse event: Not reported. No details of assessment reported

Nervous system disorders‐related AE: Not reported

Psychiatric disorders‐related AE: Not reported

Notes

Funding: Diabetes UK grant

Conflicts of interest: The authors declared that they have no conflicts of interest relevant to the study.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No details reported

Allocation concealment (selection bias)

Unclear risk

No details reported

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

No details reported

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No details reported

Incomplete outcome data (attrition bias)
All outcomes

High risk

One patient excluded from ITT‐analysis

Selective reporting (reporting bias)

High risk

Tolerability and safety outcomes not reported

Group similarity at baseline

Low risk

Similar demographic and clinical characteristics at baseline

Sample size bias

High risk

< 50 participants per treatment arm

Serpell 2014

Study characteristics

Methods

Disease: post‐herpetic neuralgia, peripheral neuropathy, focal nerve lesion, radiculopathy or CRPS type 2 associated with allodynia

Study setting: 21 centres in the UK, 7 centres in Czech Republic, 6 centres in Romania, 4 centres in Belgium 1 one centre in Canada; 2005‐2006

Study design: parallel

Study duration: 15‐week (1‐week baseline and 14‐week treatment period)

Participants

Inclusion criteria: age ≥18 years, mechanical allodynia within the territory of the affected nerve(s) (confirmed by either a positive response to stroking the allodynic area with a SENSELABTM Brush 05 (Somedic AB, Hörby, Sweden) or to force applied by a 5.07 g Semmes‐Weinstein monofilament), at least a 6‐month disease history (post‐herpetic neuralgia, peripheral neuropathy, focal nerve lesion, radiculopathy or CRPS CRPS type 2), receiving the appropriate treatment, sum score of at least 24 on a pain 0–10 NRS for more than 6 days (baseline days 2–7) during the baseline period (average 0–10 NRS score of 4/10), and pain that was not wholly relieved by their current therapy. Stable analgesic regimen for at least 2 weeks preceding study entry.

Exclusion criteria: severe pain from other concomitant conditions; history of significant psychiatric, renal, hepatic, cardiovascular or convulsive disorders, or with a known hypersensitivity to the study medication; CRPS type 1, cancer‐related PNP or pain resulting from diabetes mellitus; receiving a prohibited medication (including cannabis or cannabinoid‐based medications (in the last year), any analgesics taken on a ‘PRN’ (when required) basis, the introduction of any new analgesic medication, or any alteration to the dosage of the patient’s concomitant analgesic medication (other than the rescue analgesia provided), or all paracetamol‐containing medications (stopped on the day the patient entered the baseline period)), patients unwilling to abstain for the study duration; patients with a known history of alcohol or substance abuse; women of child‐bearing potential or their partners unless willing to ensure effective contraception was used throughout the study, participants who had received an investigational medicinal product within 12 weeks of screening; pregnant or lactating women and those planning a pregnancy; people with any physical abnormality at screening (i.e. any abnormalities that, in the opinion of the investigator, would prevent the participant from safely participating in the study), or those intending to travel or donate blood during the study

Treatment group (delta‐9‐THC): N = 128; 66% female; mean age 57.6 (mean age 14.4) years; 99% white; duration of neuropathic pain 6.3 (SD 6.7 years), 13 with cannabis exposure (10%)

Placebo group: N = 118, 55% female; mean age 57 (SD 14.1) years; 98% white; duration of neuropathic pain 6.3 (SD 6.4) years, 12 with cannabis exposure (10%)

Interventions

Study medication: pump action oromucosal spray, each 100 μL spray of THC/CBD delivered 2.7 mg of THC and 2.5 mg of CBD, each spray of placebo delivered the excipients plus colorants, both THC/CBD spray and placebo contained peppermint oil to blind the smell and taste, maximum of eight sprays in a 3‐h period up to a maximum of 24 sprays per 24‐h period

Rescue medication: paracetamol 500 mg, max. Single dose 1 g, max. Daily dose 4 g

Allowed co‐therapies: concomitant analgesic medication, with the exception of paracetamol (acetaminophen), provided that a stable dose was maintained throughout the study

Outcomes

Participant‐reported pain relief ≥ 50%: NRS 0‐10. Only OR reported: not suited for meta‐analysis (P = 0.157)

PGIC much or very much improved: reported

Withdrawal due to AE: reported

Serious AE: reported; systematic assessment

Participant‐reported pain relief ≥ 30%: NRS 0‐10; only OR reported: not suited for meta‐analysis (P = 0.021)

Mean pain intensity: Neuropathic pain scale: data not suited for meta‐analysis (P = 0.069)

HRQoL: EQ‐5D Health Status 100 to 0

Sleep problems: sleep quality 10‐0; SD calculated from P value

Fatigue: not assessed

Psychological distress: General Health Questionnaire 0‐48: SD calculated from P value

Withdrawals due to lack of efficacy: reported

Any adverse event: reported; "systematic assessment"

Nervous system disorders‐related AE: reported; systematic assessment

Psychiatric disorders‐related AE: reported; systematic assessment

Notes

Funding: GW Pharmaceuticals. GW Pharmaceuticals was involved in the study design, data collection and analysis, as well as in the preparation of this manuscript and publication decisions

Conflicts of interest: all authors received investigator fees from GW Pharma Ltd (GW)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated randomisation code

Allocation concealment (selection bias)

Low risk

Treatment allocation by GW Biometrics department; sealed code break envelopes for each partcipant

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

THC/CBD and placebo spray contained peppermint oil to blind to taste and smell

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No details reported

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

ITT by LOCF

Selective reporting (reporting bias)

Low risk

Study protocol (NCT 00710554) available; all predefined outcomes reported

Group similarity at baseline

Low risk

Similar demographic and clinical characteristics at baseline

Sample size bias

Unclear risk

50‐199 participants per treatment arm

Svendsen 2004

Study characteristics

Methods

Disease: MS (central pain)

Study setting: outpatient clinic, University Hospital of Aarhus, Denmark; study period 2001

Study design: cross‐over

Study duration: 15‐20 days with washout period of at least 21 days (actually 19‐57), 1 week baseline, 3 weeks intervention, 3 weeks washout, 3 weeks intervention

Participants

Inclusion criteria: diagnosed with MS, aged 18‐55 years, pain ≥ 3 on 0‐10 NRS, investigators assessed central pain examination, central pain being a pain in a body territory with abnormal sensation to pinprick, touch, warmth, cold, ability to differentiate central from spasticity‐related pain

Exclusion criteria: musculoskeletal disorders, PNP, visceral pain at max. pain site, hypersensitivity to cannabis‐based medicines or sesame oil, heart disease, mania, depression or schizophrenia, alcohol or drug misuse, no antidepressants, anticholinergic, antihistaminic agents or CNS depressants, use of analgesic drugs, (medications had to be stopped 1 week before first visit) pregnancy or lactation, sexually active women without reliable contraception, other clinical trials, lack of co‐operation, use of marijuana within 3 months before the study, unwillingness to abstain from marijuana use

Treatment group (dronabinol) and placebo group: N = 24; 41.7% male, mean age 50 (23‐55) years, no ethnic group, current cannabis use not reported

Interventions

Study medication: dronabinol starting with 1 x 2.5 mg capsules up to 2 x 5 mg/d

Rescue medication: paracetamol

Allowed co‐therapies: spasmolytic drugs and paracetamol

Outcomes

Participant‐reported pain relief ≥ 50%: reported. NRS 0‐10 (end of treatment period)

PGIC much or very much improved: not assessed

Withdrawal due to AE: reported

Serious AE: reported

Participant‐reported pain relief ≥ 30%: not reported.Not calculable by imputation method because baseline values not reported

Mean pain intensity: median spontaneous pain intensity NRS 0‐10 during the last week of treatment

HRQoL: SF‐36 physical functioning (50‐0); data of first treatment period used for analysis; SD calculated from P value

Sleep problems: not assessed

Fatigue: not assessed

Psychological distress: SF‐36 mental health (50‐0). Data of first treatment period used for analysis; SD calculated from P value

Withdrawals due to lack of efficacy: reported

Any adverse event: reported. "Patient used their own words to record AEs in diaries"

Nervous system disorders‐related AE: reported

Psychiatric disorders‐related AE: reported

Notes

Funding: the study was supported by grants from the Danish Multiple Sclerosis Society (grant no 2002/71045), grant 900035 from manager Ejnar Jonasseon and his wife’s memorial grant, and the Warwara Larsen Foundation (grant no 664.28), Denmark. Solvay Pharmaceuticals provided study medication (dronabinol (Marinol) and placebo capsules), labelling, and packaging. In addition, the company provided financial support for study monitoring and data analysis. IPC‐Nordic, Denmark, packaged and labelled the study medication and monitored the study. These companies were not involved in the design or execution of the study or writing the manuscript.

Conflicts of interest: none declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"We assigned patients to treatment sequence by using a computer generated randomisation code with a block size of six prepared by IPC‐Nordic"

Allocation concealment (selection bias)

Low risk

"Investigators allocated patients consecutively by time of inclusion at the study site. One investigator (KBS) enrolled all participants and allocated them to treatment".

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"We administered both active treatment and placebo as white capsules (soft gelatin capsules) in identical containers. The taste and smell of the capsules did not differ."

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No details provided

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All participants terminated the study

Selective reporting (reporting bias)

Unclear risk

No study protocol reported

Group similarity at baseline

Low risk

No significant differences in demographic and clinical characteristics between the study groups because of study design

Sample size bias

High risk

< 25 participants per treatment arm

Toth 2012

Study characteristics

Methods

Disease: diabetic peripheral polyneuropathy

Study setting: single‐centre, Canada; study period not reported

Study design: EERW

Study duration: single‐blind for 4 weeks, double‐blind randomised withdrawal for 5 weeks

Participants

Inclusion criteria: DPN pain questionnaire score ≥ 4, pain duration at least 3 months, pain severity with averaged scores of P40 mm on the 100‐mm VAS of the short‐Form McGill Pain Questionnaire

Exclusion criteria: participants with other causes of pain, including PHN, lumbar radiculopathy, central neuropathic pain, CRPSs I or II, or significant osteoarthritis, were excluded. Any skin conditions over the area of DPN which could hinder examination, led to exclusion. Any current diagnoses of schizophrenia, psychotic disorder, bipolar affective disorder, obsessive compulsive disorder, or major depressive disorder were also exclusionary. Clinically significant unstable medical conditions that could compromise participation, such as with poor diabetic control (haemoglobin A1C ≥ 11%), history of substance abuse or dependence, malignancy other than squamous cell carcinoma in the last 2 years, elevation of liver enzymes above 3 times the upper limit of normal, or an anticipated need for surgery or hospitalisation within the next 16 weeks after screening led to exclusion at the discretion of the investigator. Those participants previously exposed to nabilone were excluded. Any use of self‐obtained cannabis‐based medicines or other illicit drugs during the study was prohibited, and participants with a positive urinary illicit drug screen (including detection of 11‐nor‐delta‐9‐ tetrahydrocannabinol‐9‐carboxylic acid) were excluded at screening.

Treatment group (nabilone (delta‐9‐THC)): N = 13; mean age 61.6 (SD 14.6) years; 69% male; 92% white; duration of diabetes 10 (SD 12.6) years. No reports on previous cannabis use

Placebo group: N = 13; mean age 60.8 (SD 15.2) years; 38% male; 92% white; duration of diabetes 9.7 (SD 13.1) years. No reports on previous cannabis use

Interventions

Study medication: nabilone 1 mg‐5 mg/d orally

Rescue medication: placebo drug

Allowed co‐therapies: no details provided

Outcomes

Participant‐reported pain relief ≥ 50%: reported (NRS 0‐10 over the preceding 24 h)

PGIC much or very much improved: reported (in figure)

Withdrawal due to AE: reported

Serious AE: reported

Participant‐reported pain relief ≥ 30%: reported

Mean pain intensity: average pain intensity (VAS 0‐10)

HRQoL: Euro‐QOL VAS 100‐0

Sleep problems: Medical Outcomes Study Sleep problems index: reported

Fatigue: not assessed

Withdrawals due to lack of efficacy: reported

Any adverse event: reported; "All spontaneously reported and observed AEs were recorded at each clinic visit and during telephone follow‐up visits"

Nervous system disorders‐related AE: incompletely reported. Not suited for meta‐analysis

Psychiatric disorders‐related AE: reported

Notes

Funding: Valeant

Conflicts of interest: Dr. Toth received honoraria from Valeant Canada for educational lectures.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Electronic randomization system was used to randomise individual subjects without block randomisation as developed by an outside coordinator"

Allocation concealment (selection bias)

Low risk

"Randomization was concealed from subjects, clinical coordinator, and assessing physicians"

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"Medication was blinded for placebo using capsules of identical size, colour, taste, and smell."

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No details reported

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

ITT by LOCF

Selective reporting (reporting bias)

Unclear risk

Study protocol available (NCT01035281) but no outcomes reported

Group similarity at baseline

High risk

Significant difference in sex ratio at baseline

Sample size bias

High risk

< 25 participants per treatment arm

Ware 2010

Study characteristics

Methods

Disease: non HIV neuropathy > 3 months duration caused by trauma, surgery; with pain ≥ 40/100 VAS, stable analgesic regimen

Study setting: single‐centre university, Canada; 2003‐2006

Study design: 4 periods cross‐over

Study duration: 2 weeks with 5 treatment days per each period, 9 days' washout

Participants

Inclusion criteria: men and women aged ≥ 18 years with neuropathic pain of at least 3 months in duration caused by trauma or surgery, with allodynia or hyperalgesia, and with an average weekly pain intensity score > 4 on a 10‐cm VAS. Participants had a stable analgesic regimen and reported not having used cannabis during the year before the study Potential participants had to have normal liver function (defined as aspartate aminotransferase < 3 times normal), normal renal function (defined as a serum creatinine level < 133 μmol/L), normal haematocrit (> 38%) and a negative result on β human chorionic gonadotropin pregnancy test (if applicable).

Exclusion criteria: pain due to cancer or nociceptive causes, presence of significant cardiac or pulmonary disease, current substance abuse or dependence (including abuse of or dependence on cannabis), history of psychotic disorder, current suicidal ideation, pregnancy or breastfeeding, participation in another clinical trial within 30 days of enrolment, and ongoing insurance claims

Treatment group (delta‐9‐THC)/placebo group): N = 23 participants, mean age: 45.4 years (SD 12.3); gender (male/female): 11/12; 18 (81%) with previous cannabis exposure, but not within the year prior to the study

Interventions

Study medication: 3 different potencies of THC (2.5%, 6%, 9.4%) from whole herb in gelatine capsules inhaled through pipe. Placebo cigarettes underwent ethanolic extraction. Dose estimate: 0, 1.625, 3.9 and 5.85 mg/d (average) THC per period

Rescue medication: not reported

Allowed co‐therapies: "Stable regimen"

Outcomes

Participant‐reported pain relief ≥ 50%: not reported and not calculable by imputation method. Average daily pain Intensity on 0‐10 NRS average over 5 treatment days

PGIC much or very much improved: not assessed

Withdrawal due to AE: reported

Serious AE attributed to study medication: reported

Participant‐reported pain relief ≥ 30%: not reported and not calculable by imputation method

Mean pain intensity: average daily pain intensity on 0‐10 NRS

HRQoL: EQ‐5D state of health VAS 100‐0

Sleep problems: sleep quality Leeds Sleep Evaluation Questionnaire 0‐10

Fatigue: not assessed

Psychological distress: Profile of Mood States total mood disturbance 0‐200

Withdrawals due to lack of efficacy: not reported

Any adverse event: reported; No details of assessment reported

Nervous system disorders‐related AE: reported

Psychiatric disorders‐related AE: reported

Notes

Funding: Canadian Institutes of Health (JHM 50014) and Louise and Alan Wards Foundation

Conflicts of interest: the study authors declare that they have not conflict of interest.

"We found no evidence of significant carry‐over effect for any outcome"

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No details reported

Allocation concealment (selection bias)

Unclear risk

No details for investigators reported. Participants correctly guessed allocation at the end of the trial

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

No details reported

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No details reported

Incomplete outcome data (attrition bias)
All outcomes

High risk

No ITT

Selective reporting (reporting bias)

Low risk

Consistent reporting according to study protocol (ISRCT68314063)

Group similarity at baseline

Low risk

Identical demographic and baseline characteristics due to study protocol

Sample size bias

High risk

< 25 participants per treatment arm

AE: adverse events; bpm: beats per minute; BSI: Brief Symptom Inventory; CBD: cannabidiol; CNS: central nervous system; CRPS: complex regional pain syndrome; DHC: dihydrocodeine; DPN: diabetic peripheral neuropathic; DSM: Diagnostic and Statistical Manual of Mental Disorders; EERW: enriched enrolment randomised withdrawal; EQ‐5D: EuroQol quality of life instrument; HR: heart rate; HRQoL: Health‐related quality of life; ITT: intention‐to‐treat; LOCF: last observation carried forward; mg: milligrams; MAO: monoamine oxidase; MI: myocardial infarction; μL = microlitre; mL = millilitre; µmol/L: micromoles per litre; MS: multiple sclerosis; N: number; NDS: Neuropathy Disability Score; NNTB: number needed to treat for an additional beneficial outcome; NRS: numerical rating scale; NSAIDs: non‐steroidal anti‐inflammatory drugs; OR: odds ratio; PGIC: Patient Global Impression of Change; PHN: postherpetic neuralgia; PNP: peripheral neuropathic pain; SD; standard deviation; SIP: Sickness Impact Profile;SF‐36: short‐form 36 quality of life instrument; TENS: transcutaneous electrical nerve stimulation; THC: tetrahydrocannabinol; VAS: visual analogue scale; WHO: World Health Organization

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Abrams 2007

Cannabis cigarettes or placebo cigarettes in 55 participants. HIV‐associated neuropathy; study duration < 2 weeks

Corey‐Bloom 2012

Smoked cannabis or placebo cigarettes in 30 participants with MS for 2 weeks; no definite statement that the pain was of neuropathic nature

Karst 2003

Synthetic THC or oral placebo in 21 participants with chronic neuropathic central and peripheral pain of various aetiologies; study duration < 2 weeks

Notcutt 2011

34 ‘N of 1’ studies with THC, CBD and THC/CBD or placebo over 12 weeks; 2 participants with non‐neuropathic pain included

Novotna 2011

572 participants with MS were treated with THC/CBD spray for 12 weeks; participants were selected because of spasticity refractory to conventional treatment; no definite statement that the pain was of neuropathic nature

Rintala 2010

Randomised, controlled, double‐blind, cross‐over pilot study with 7 participants with spinal cord injury and neuropathic pain comparing dronabinol with diphenhydramine; < 10 participants per treatment arm

Turcotte 2015

15 participants with MS‐induced neuropathic pain were treated with nabilone as an adjunctive to gabapentin for 9 weeks; < 10 participants per treatment arm

Wade 2003

20 participants with neurogenic symptoms due to lesions of the central or peripheral nervous system were treated with plant‐based THC/CBD for 2 weeks in a cross‐over design. 13 of 20 participants with pain. No statement or analysis that carry‐over effects were excluded

Wade 2004

160 participants with MS treated with THC/CBD spray or placebo spray of 6 weeks; no definite statement that the pain was of neuropathic nature

Wallace 2015

Inhaled cannabis in 16 participants with painful diabetic polyneuropathy for 4 single dosing sessions. Study duration < 2 weeks

Wilsey 2008

Vaporised cannabis (1.3% and 3.5%) or placebo in 39 participants with central and peripheral neuropathic pain for 1 day (experimental study)

Wilsey 2013

38 participants with central or peripheral neuropathic pain were treated with smoked cannabis or placebo. Study duration < 1 week

Wissel 2006

Nabilone or placebo in 11 participants with MS und upper motor neuron disease‐associated spasticity‐related pain for 4 weeks; no definite statement that the pain was of neuropathic nature

Zajicek 2003

667 participants with MS were treated with oral cannabis extract (THC) or delta 9‐THC or placebo for 15 weeks. Spasticity was the primary outcome. Pain was a secondary outcome; only around 65% of participants had pain, with no pain intensity at baseline reported

Zajicek 2012

275 patients with MS were treated for 12 weeks with plant‐derived THC 2.5‐15 mg/d orally or placebo. No definite statement that the pain was of neuropathic nature

CBD: cannabidiol; mg: milligrams; µmol/L: micromoles per litre;MS: multiple sclerosis; THC: tetrahydrocannabinol;

Characteristics of studies awaiting classification [ordered by study ID]

NCT00699634

Methods

Disease: phantom limb pain

Study setting: single‐centre university, Canada; 2009‐2011

Study design: parallel

Study duration: 6 weeks

Participants

Inclusion criteria

  1. Diagnosed with phantom limb pain by a Rehabilitation Medicine Specialist

  2. 18‐70 years old

  3. Any gender

  4. No resolution of phantom limb pain with other treatments, such as a tricyclic antidepressant, or anticonvulsant medication

  5. No previous use of oral cannabis‐based medicines for pain management

Exclusion criteria:

  1. Pain is better explained by a treatable cause of stump pain, such as neuroma or bony overgrowth

  2. Gross abnormalities on routine baseline blood work including electrolytes, urea and creatinine, a complete blood count, and liver function tests (AST ALT GGT, Alk Phos, and LDH) that are twice the limit of normal. Normal tests taken within 3 months prior to the study accepted if there is no history of acute illness since the time the blood was drawn.

  3. Heart disease. (Cannabis‐based medicines can reduce heart rate and blood pressure). People with heart disease excluded based on a history of symptomatic angina, MI or congestive heart failure as well as a clinical exam.

  4. Schizophrenia or other psychotic disorder

  5. Severe liver dysfunction

  6. History of untreated non‐psychotic emotional disorders

  7. Cognitive impairment

  8. Major illness in another body area

  9. Pregnancy

  10. Nursing mothers

  11. History of drug dependency

  12. Known sensitivity to marijuana or other cannabinoid agents.

Treatment group nabilone/placebo group: N = not reported

Interventions

Study medication: nabilone 0.5 mg at bedtime for 1 week, then 0.5 mg twice daily for 1 week. After a reassessment of the outcome measures, the dose is increased to 0.5 mg in the morning and 1 mg at hs for 1 week, followed by an increase to 1 mg twice daily in the last week of the study.

Rescue medication: not reported

Allowed co‐therapies: not reported

Outcomes

Participant‐reported pain relief ≥ 50%: not reported

PGIC much or very much improved: not assessed

Withdrawal due to AE: not reported

Serious AE attributed to study medication: not reported

Participant‐reported pain relief ≥ 30%: not reported

Mean pain intensity: VAS for pain; not reported

HRQoL:: SF‐36 not reported

Sleep problems: Groningen Sleep Quality Scale; not reported

Fatigue: not assessed

Psychological distress: Hospital Anxiety and Depression Scale not reported

Withdrawals due to lack of efficacy: not reported

Any adverse event: reported; no details of assessment reported

Nervous system disorders‐related AE: not reported

Psychiatric disorders‐related AE: not reported

Notes

Funding: Valeant, University of Manitoba

Conflicts of interest: not declared

NCT01035281

Methods

Disease: diabetic neuropathic pain

Study setting: single‐centre university, Canada; start 2009; the recruitment status of this study is unknown because the information has not been verified recently.

Study design: EERW

Study duration: all participants who experienced at least a 30% reduction in their weekly mean pain score during the 4‐week, single‐blind flexible dosing phase considered a responder, and further continued in the study. During the double‐blind portion of the study, participants randomised to nabilone continued on the dose of nabilone achieved at the completion of the single‐blind phase, and this dose was maintained throughout the double‐blind phase. Participants randomised to placebo received 1 mg of nabilone daily for 1 week, followed by 4 consecutive weeks of placebo. This dose of nabilone permitted a tapering for those participants achieving a higher daily dose of nabilone during the single‐blind phase, or maintained those who were taking only 1 mg/d in the single‐blind phase, preventing an abrupt termination of treatment in participants who were randomised into the placebo portion.

Participants

Inclusion criteria:

  1. Male or female participants, aged 18‐80 years

  2. Signed and dated informed consent

  3. Women of childbearing potential had to have a negative serum β‐HCG pregnancy test and be practicing an effective form of contraception (accepted methods are hormonal (oral contraceptive or injectable contraceptive), double barrier with spermicide, or intrauterine device‐IUD). Complete abstinence may be considered acceptable, but must be determined on a case‐by‐case basis with the clinical investigator.

  4. Diagnosis of DPN‐associated neuropathic pain syndrome, confirmed by a qualified neurologist or pain specialist, with persistence for a minimum of 3 months

  5. Score of ≥ 4 on the Douleur Neuropathique 4 (DN4) questionnaire, a single‐page survey consisting of historical questions and 1 examination portion using light touch and pinprick over the region of suspected neuropathic pain. This has high sensitivity and specificity for neuropathic pain

  6. Must complete ≥ 4 daily pain diaries during the week of the screening phase prior to randomisation

  7. Must have a daily mean pain score of ≥ 4 over the screening period prior to randomisation based on Daily Pain Rating Scale (DPRS).

  8. Must have a score of > 40 mm on the VAS of the Short Form McGill Pain Questionnaire (SF‐MPQ).

  9. Screening laboratory values must be within normal limits, or abnormalities must be deemed clinically insignificant in the judgment of the investigator

  10. Participant must be deemed capable of complying with study schedule, procedures and medications

Exclusion criteria:

  1. Pregnant or lactating women or women of childbearing potential not using acceptable method of contraception

  2. Participants with neuropathic pain that is not due to DPN

  3. Any skin conditions in the affected areas with NeP that (in the judgment of the investigator) could interfere with evaluation of the NeP

  4. Current or past DSM‐IV‐TR (Text Revision)(2000) diagnosis of schizophrenia, psychotic disorder, bipolar affective disorder or obsessive‐compulsive disorder and Major Depressive Disorder (MDD).

  5. Current or past DSM‐IV‐TRTM (2000) diagnosis of substance abuse or dependence within the last 6 months.

  6. Use of marijuana or other cannabis‐based medicines during the study. Discontinuation of these substances 30 days prior to the screening visit is permitted. The study consent must be signed and dated prior to the discontinuation of these substances.

  7. Clinically significant or unstable conditions that, in the opinion of the investigator, would compromise participation in the study. This includes, for example, medical conditions such as, but not limited to: hepatic, renal, respiratory, haematological, immunologic, or cardiovascular diseases (e.g. MI within previous month, ventricular arrhythmia recent severe heart insufficiency), inflammatory or rheumatologic disease, active infections, symptomatic peripheral vascular disease, and untreated endocrine disorders

  8. History of seizure disorder, except febrile seizures of childhood

  9. A glycated haemoglobin (HbA1C) of > 11% at screening

  10. Any other condition, which in the investigator's judgment might increase the risk to the participant or decrease the chance of obtaining satisfactory data to achieve the objectives of the study. This includes any condition precluding nabilone use.

  11. Malignancy within past 2 years with exception of basal cell carcinoma

  12. Urine screen positive for illicit substances, including THC such as marijuana at screening (Visit 1)

  13. Liver function tests or liver enzymes > 3 times the upper limit of normal (ULN)

  14. Other blood or urine laboratory results which are sufficiently abnormal in the view of the investigator(s) to raise concern about the enrolment of this subject in this study

  15. A previous history of intolerance or hypersensitivity to cannabis‐based medicines or other medications or substances with similar chemical structure

  16. Anticipated need for surgery during the study or within 4 weeks of completion

  17. Anticipated need for general anesthetics during the course of the study

  18. Anticipated need for hospitalisation for any reason during the course of the study or within 4 weeks of completion

  19. Previous prescribed use of nabilone or other cannabis‐based medicines, including use of sample medications, within the 30 days prior to screening. Note that prior use of marijuana not an exclusion criterion

  20. Participation in any other studies involving investigational or marketed products, concomitantly or within 30 days prior to entry in the study and/or

  21. Employees or relatives of employees of the investigational site or Valeant Canada

Interventions

Study medication:nabilone, flexible dosing nabilone at 0.5 mg‐4 mg/d

Rescue medication: not reported

Allowed co‐therapies: not reported

Outcomes

Participant‐reported pain relief ≥ 50%: no information provided

PGIC much or very much improved: no information provided

Withdrawal due to AE: no information provided

Serious AE attributed to study medication: no information provided

Participant‐reported pain relief ≥ 30%: no information provided

Mean pain intensity: no information provided

HRQoL:: no information provided

Sleep problems: no information provided

Fatigue: no information provided

Psychological distress: no information provided

Withdrawals due to lack of efficacy: no information provided

Any adverse event: no information provided

Nervous system disorders‐related AE: no information provided

Psychiatric disorders‐related AE: no information provided

Notes

Funding: University of Calgary

Conflicts of interest: not declared

NCT01222468

Methods

Disease: neuropathic pain in spinal cord injured persons

Study setting: single‐centre university, Canada; 2012‐2015

Study design: cross‐over

Study duration: 11 weeks each period

Participants

Inclusion criteria

  1. Spinal Cord Injury

  2. 12 months post ‐injury

  3. Cervical spine 2‐Thoracic spine 12, ASIA Impairment scale categories A‐D, stable level of injury

  4. Moderate‐severe spasticity or moderate to severe neuropathic pain

  5. No cognitive impairment

  6. Spasticity medications unchanged for at least 30 days or inadequate pain control at a stabilised dose of either gabapentin or pregabalin for at least 30 days

  7. No botulinum toxin injections x 6 months

Exclusion criteria

  1. Significant cardiovascular disease

  2. Major illness in another body area

  3. History of psychological disorders or predisposition to psychosis

  4. Sensitivity to cannabis‐based medicines

  5. Severe liver dysfunction

  6. History of drug dependency

  7. Fixed tendon contractures

  8. Used cannabis in the past 30 days

  9. Unwilling to refrain from smoking cannabis during the study

  10. Pregnant or nursing mother

Treatment group nabilone/placebo group: N = not reported

Interventions

Study medication: nabilone 0.5 mg tablets od titrated to a maximum daily dose of 3 mg by mouth over an 11‐week phase; placebo 0.5 mg by mouth daily, dose titrated to a maximum daily dose of 3.0 mg by mouth over an 11‐week phase

Rescue medication: not reported

Allowed co‐therapies: not reported

Outcomes

Participant‐reported pain relief ≥ 50%: not reported

PGIC much or very much improved: not reported

Withdrawal due to AE: not reported

Serious AE attributed to study medication: not reported

Participant‐reported pain relief ≥ 30%: not reported

Mean pain intensity: VAS for pain and Neuropathic Pain Questionnaire; not reported

HRQoL:: SF‐36 not reported

Sleep problems: Pittsburgh Sleep Quality Index; not reported

Fatigue: not assessed

Psychological distress: not assessed

Withdrawals due to lack of efficacy: not reported

Any adverse event: reported; no details of assessment reported

Nervous system disorders‐related AE: not reported

Psychiatric disorders‐related AE: not reported

Notes

Funding: University of Manitoba The Manitoba Spinal Cord Injury Research Fund Canadian Paraplegic Association Health Sciences Centre Foundation, Manitoba

Conflicts of interest: not declared

AE; adverse events; ALT: alanine aminotransferase; AST: aspartate aminotransferase; DSM: Diagnostic and Statistical Manual of Mental Disorders; EERW: enriched enrolment randomised withdrawal; GGT: gamma‐glutamyl transferase; HRQoL: health‐related quality of life; mg: milligrams; MI: myocardial infarction; N: number; PGIC: Patient Global Impression of Change; SF‐36: short‐form 36 quality of life instrument; THC: tetrahydrocannabinol; VAS: visual analogue scale

Data and analyses

Open in table viewer
Comparison 1. Cannabis‐based medicines versus placebo at final treatment

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1.1 Pain relief of 50% or greater Show forest plot

8

1001

Risk Difference (IV, Random, 95% CI)

0.05 [0.00, 0.09]

Analysis 1.1

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 1: Pain relief of 50% or greater

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 1: Pain relief of 50% or greater

1.1.1 Central pain ‐ multiple sclerosis

4

669

Risk Difference (IV, Random, 95% CI)

0.08 [‐0.00, 0.15]

1.1.2 Peripheral pain ‐ chemotherapy‐induced polyneuropathy

1

36

Risk Difference (IV, Random, 95% CI)

0.11 [‐0.06, 0.28]

1.1.3 Peripheral pain ‐ diabetic polyneuropathy

1

30

Risk Difference (IV, Random, 95% CI)

‐0.20 [‐0.54, 0.14]

1.1.4 Peripheral pain ‐ plexus injury

1

141

Risk Difference (IV, Random, 95% CI)

0.01 [‐0.04, 0.06]

1.1.5 Peripheral pain ‐ polyneuropathy of various aetiologies

1

125

Risk Difference (IV, Random, 95% CI)

0.13 [0.00, 0.25]

1.2 Patient Global Impression much or very much improved Show forest plot

6

1092

Risk Difference (IV, Random, 95% CI)

0.09 [0.01, 0.17]

Analysis 1.2

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 2: Patient Global Impression much or very much improved

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 2: Patient Global Impression much or very much improved

1.2.1 Central pain ‐ multiple sclerosis

2

397

Risk Difference (IV, Random, 95% CI)

0.06 [‐0.01, 0.14]

1.2.2 Central pain ‐ spinal cord injury

1

116

Risk Difference (IV, Random, 95% CI)

0.34 [0.17, 0.50]

1.2.3 Peripheral pain ‐ diabetic polyneuropathy

1

281

Risk Difference (IV, Random, 95% CI)

0.02 [‐0.09, 0.14]

1.2.4 Peripheral pain ‐ polyneuropathy of various aetiologies

1

228

Risk Difference (IV, Random, 95% CI)

0.08 [‐0.02, 0.17]

1.2.5 Central or peripheral pain ‐ various aetiologies

1

70

Risk Difference (IV, Random, 95% CI)

‐0.01 [‐0.22, 0.19]

1.3 Withdrawals due to adverse events Show forest plot

13

1848

Risk Difference (IV, Random, 95% CI)

0.04 [0.02, 0.07]

Analysis 1.3

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 3: Withdrawals due to adverse events

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 3: Withdrawals due to adverse events

1.3.1 Central pain ‐ multiple sclerosis

4

693

Risk Difference (IV, Random, 95% CI)

0.04 [0.01, 0.08]

1.3.2 Central pain ‐ spinal cord injury

1

116

Risk Difference (IV, Random, 95% CI)

0.09 [0.01, 0.17]

1.3.3 Peripheral pain ‐ chemotherapy‐induced polyneuropathy

1

36

Risk Difference (IV, Random, 95% CI)

0.00 [‐0.10, 0.10]

1.3.4 Peripheral pain ‐ diabetic polyneuropathy

1

297

Risk Difference (IV, Random, 95% CI)

0.12 [0.04, 0.20]

1.3.5 Peripheral pain ‐ HIV polyneuropathy

1

68

Risk Difference (IV, Random, 95% CI)

0.00 [‐0.13, 0.13]

1.3.6 Peripheral pain ‐ plexus injury

1

141

Risk Difference (IV, Random, 95% CI)

0.01 [‐0.04, 0.06]

1.3.7 Peripheral pain ‐ polyneuropathy of various aetiologies

3

427

Risk Difference (IV, Random, 95% CI)

0.08 [0.02, 0.13]

1.3.8 Central and peripheral pain ‐ various aetiologies

1

70

Risk Difference (IV, Random, 95% CI)

‐0.06 [‐0.19, 0.07]

1.4 Serious adverse events Show forest plot

13

1876

Risk Difference (IV, Random, 95% CI)

0.01 [‐0.01, 0.03]

Analysis 1.4

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 4: Serious adverse events

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 4: Serious adverse events

1.4.1 Central pain ‐ multiple sclerosis

4

693

Risk Difference (IV, Random, 95% CI)

0.03 [‐0.01, 0.06]

1.4.2 Central pain ‐ spinal cord injury

1

116

Risk Difference (IV, Random, 95% CI)

0.02 [‐0.05, 0.09]

1.4.3 Peripheral pain ‐ chemotherapy‐induced neuropathy

1

36

Risk Difference (IV, Random, 95% CI)

0.00 [‐0.10, 0.10]

1.4.4 Peripheral pain ‐ diabetic polyneuropathy

1

297

Risk Difference (IV, Random, 95% CI)

0.01 [‐0.05, 0.08]

1.4.5 Peripheral pain ‐ HIV polyneuropathy

1

68

Risk Difference (IV, Random, 95% CI)

0.03 [‐0.07, 0.13]

1.4.6 Peripheral pain ‐ plexus injury

1

141

Risk Difference (IV, Random, 95% CI)

0.00 [‐0.03, 0.03]

1.4.7 Peripheral pain ‐ polyneuropathies of various aetiologies

3

455

Risk Difference (IV, Random, 95% CI)

0.01 [‐0.02, 0.04]

1.4.8 Central and peripheral pain ‐ various aetiologies

1

70

Risk Difference (IV, Random, 95% CI)

‐0.06 [‐0.15, 0.03]

1.5 Pain relief of 30% or greater Show forest plot

10

1586

Risk Difference (IV, Random, 95% CI)

0.09 [0.03, 0.15]

Analysis 1.5

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 5: Pain relief of 30% or greater

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 5: Pain relief of 30% or greater

1.5.1 Central pain ‐ multiple sclerosis

3

645

Risk Difference (IV, Random, 95% CI)

0.11 [‐0.03, 0.25]

1.5.2 Peripheral pain ‐ chemotherapy‐induced polyneuropathy

1

36

Risk Difference (IV, Random, 95% CI)

0.11 [‐0.16, 0.38]

1.5.3 Peripheral pain ‐ diabetic polyneuropathy

2

327

Risk Difference (IV, Random, 95% CI)

‐0.04 [‐0.14, 0.07]

1.5.4 Peripheral pain ‐ HIV polyneuropathy

1

56

Risk Difference (IV, Random, 95% CI)

0.29 [0.05, 0.52]

1.5.5 Peripheral pain ‐ plexus injury

1

141

Risk Difference (IV, Random, 95% CI)

0.10 [‐0.06, 0.25]

1.5.6 Peripheral pain ‐ polyneuropathy of various aetiologies

2

381

Risk Difference (IV, Random, 95% CI)

0.11 [0.03, 0.19]

1.6 Mean pain intensity Show forest plot

14

1837

Std. Mean Difference (IV, Random, 95% CI)

‐0.35 [‐0.60, ‐0.09]

Analysis 1.6

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 6: Mean pain intensity

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 6: Mean pain intensity

1.6.1 Central pain ‐ multiple sclerosis

4

668

Std. Mean Difference (IV, Random, 95% CI)

‐0.10 [‐0.25, 0.05]

1.6.2 Central pain ‐ spinal cord injury

1

114

Std. Mean Difference (IV, Random, 95% CI)

‐0.04 [‐0.41, 0.33]

1.6.3 Peripheral pain ‐ chemotherapy‐induced polyneuropathy

1

36

Std. Mean Difference (IV, Random, 95% CI)

‐0.20 [‐0.86, 0.45]

1.6.4 Peripheral pain ‐ diabetic polyneuropathy

2

324

Std. Mean Difference (IV, Random, 95% CI)

‐0.05 [‐0.27, 0.17]

1.6.5 Peripheral pain ‐ HIV polyneuropathy

1

56

Std. Mean Difference (IV, Random, 95% CI)

‐0.41 [‐0.94, 0.12]

1.6.6 Peripheral pain ‐ plexus injury

1

141

Std. Mean Difference (IV, Random, 95% CI)

‐0.43 [‐0.79, ‐0.08]

1.6.7 Peripheral pain ‐ polyneuropathy of various aetiologies

3

428

Std. Mean Difference (IV, Random, 95% CI)

‐0.65 [‐1.75, 0.44]

1.6.8 Central and peripheral pain ‐ various aetiologies

1

70

Std. Mean Difference (IV, Random, 95% CI)

‐0.24 [‐0.71, 0.23]

1.7 Health‐related quality of life Show forest plot

9

1284

Std. Mean Difference (IV, Random, 95% CI)

0.02 [‐0.10, 0.13]

Analysis 1.7

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 7: Health‐related quality of life

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 7: Health‐related quality of life

1.7.1 Central pain ‐ multiple sclerosis

2

363

Std. Mean Difference (IV, Random, 95% CI)

‐0.07 [‐0.27, 0.14]

1.7.2 Central pain ‐ spinal cord injury

1

113

Std. Mean Difference (IV, Random, 95% CI)

0.00 [‐0.37, 0.37]

1.7.3 Peripheral pain ‐ diabetic polyneuropathy

2

303

Std. Mean Difference (IV, Random, 95% CI)

0.17 [‐0.06, 0.39]

1.7.4 Peripheral pain ‐ plexus injury

1

141

Std. Mean Difference (IV, Random, 95% CI)

‐0.07 [‐0.42, 0.28]

1.7.5 Peripheral pain of various aetiologies

2

300

Std. Mean Difference (IV, Random, 95% CI)

‐0.03 [‐0.26, 0.21]

1.7.6 Central and peripheral pain ‐ various aetiologies

1

64

Std. Mean Difference (IV, Random, 95% CI)

0.15 [‐0.35, 0.64]

1.8 Sleep problems Show forest plot

8

1386

Std. Mean Difference (IV, Random, 95% CI)

‐0.47 [‐0.90, ‐0.04]

Analysis 1.8

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 8: Sleep problems

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 8: Sleep problems

1.8.1 Central pain ‐ multiple sclerosis

1

339

Std. Mean Difference (IV, Random, 95% CI)

0.01 [‐0.21, 0.22]

1.8.2 Central pain ‐ spinal cord injury

1

114

Std. Mean Difference (IV, Random, 95% CI)

‐0.04 [‐0.41, 0.32]

1.8.3 Peripheral pain ‐ diabetic polyneuropathy

1

274

Std. Mean Difference (IV, Random, 95% CI)

‐0.14 [‐0.38, 0.10]

1.8.4 Peripheral pain ‐ plexus injury

1

141

Std. Mean Difference (IV, Random, 95% CI)

‐0.42 [‐0.78, ‐0.07]

1.8.5 Peripheral pain ‐ polyneuropathy of various aetiologies

3

448

Std. Mean Difference (IV, Random, 95% CI)

‐0.78 [‐2.17, 0.61]

1.8.6 Central and peripheral pain ‐ various aetiologies

1

70

Std. Mean Difference (IV, Random, 95% CI)

‐0.31 [‐0.78, 0.16]

1.9 Psychological distress Show forest plot

7

779

Std. Mean Difference (IV, Random, 95% CI)

‐0.32 [‐0.61, ‐0.02]

Analysis 1.9

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 9: Psychological distress

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 9: Psychological distress

1.9.1 Central pain ‐ multiple sclerosis

2

363

Std. Mean Difference (IV, Random, 95% CI)

‐0.03 [‐0.65, 0.59]

1.9.2 Peripheral pain ‐ chemotherapy‐induced polyneuropathy

1

36

Std. Mean Difference (IV, Random, 95% CI)

‐1.07 [‐1.78, ‐0.37]

1.9.3 Peripheral pain ‐ diabetic polyneuropathy

1

30

Std. Mean Difference (IV, Random, 95% CI)

‐0.25 [‐0.97, 0.47]

1.9.4 Peripheral pain ‐ plexus injury

1

141

Std. Mean Difference (IV, Random, 95% CI)

‐0.27 [‐0.62, 0.08]

1.9.5 Peripheral pain ‐ polyneuropathy of various aetiologies

2

209

Std. Mean Difference (IV, Random, 95% CI)

‐0.48 [‐0.80, ‐0.16]

1.10 Withdrawals due to lack of efficacy Show forest plot

9

1576

Risk Difference (IV, Random, 95% CI)

‐0.00 [‐0.02, 0.01]

Analysis 1.10

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 10: Withdrawals due to lack of efficacy

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 10: Withdrawals due to lack of efficacy

1.10.1 Central pain ‐ multiple sclerosis

4

697

Risk Difference (IV, Random, 95% CI)

0.00 [‐0.02, 0.02]

1.10.2 Peripheral pain ‐ diabetic polyneuropathy

1

297

Risk Difference (IV, Random, 95% CI)

‐0.01 [‐0.05, 0.03]

1.10.3 Peripheral pain ‐ plexus injury

1

141

Risk Difference (IV, Random, 95% CI)

0.00 [‐0.04, 0.04]

1.10.4 Peripheral pain ‐ polyneuropathy of various aetiologies

2

371

Risk Difference (IV, Random, 95% CI)

‐0.04 [‐0.09, 0.01]

1.10.5 Central and peripheral pain ‐ various aetiologies

1

70

Risk Difference (IV, Random, 95% CI)

0.00 [‐0.05, 0.05]

1.11 Any adverse event Show forest plot

7

1356

Risk Difference (IV, Random, 95% CI)

0.19 [0.12, 0.27]

Analysis 1.11

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 11: Any adverse event

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 11: Any adverse event

1.11.1 Central pain ‐ multiple sclerosis

3

627

Risk Difference (IV, Random, 95% CI)

0.22 [0.05, 0.39]

1.11.2 Central pain ‐ spinal cord injury

1

116

Risk Difference (IV, Random, 95% CI)

0.34 [0.18, 0.50]

1.11.3 Peripheral pain ‐ diabetic polyneuropathy

1

297

Risk Difference (IV, Random, 95% CI)

0.12 [0.02, 0.22]

1.11.4 Peripheral pain ‐ polyneuropathy of various aetiologies

1

246

Risk Difference (IV, Random, 95% CI)

0.15 [0.05, 0.25]

1.11.5 Central and peripheral pain ‐ various aetiologies

1

70

Risk Difference (IV, Random, 95% CI)

0.21 [0.06, 0.36]

1.12 Specific adverse event: nervous system disorders Show forest plot

9

1304

Risk Difference (IV, Random, 95% CI)

0.38 [0.18, 0.58]

Analysis 1.12

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 12: Specific adverse event: nervous system disorders

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 12: Specific adverse event: nervous system disorders

1.12.1 Central pain ‐ multiple sclerosis

3

453

Risk Difference (IV, Random, 95% CI)

0.33 [0.09, 0.58]

1.12.2 Central pain ‐ spinal cord injury

1

116

Risk Difference (IV, Random, 95% CI)

0.53 [0.38, 0.68]

1.12.3 Peripheral pain ‐chemotherapy‐induced polyneuropathy

1

36

Risk Difference (IV, Random, 95% CI)

1.00 [0.90, 1.10]

1.12.4 Peripheral pain ‐ diabetic polyneuropathy

1

297

Risk Difference (IV, Random, 95% CI)

0.26 [0.15, 0.37]

1.12.5 Peripheral pain ‐ polyneuropathy of various aetiologies

2

332

Risk Difference (IV, Random, 95% CI)

0.29 [0.19, 0.39]

1.12.6 Central and peripheral pain ‐ various aetiologies

1

70

Risk Difference (IV, Random, 95% CI)

0.37 [0.15, 0.58]

1.13 Specific adverse event: psychiatric disorders Show forest plot

9

1314

Risk Difference (IV, Random, 95% CI)

0.10 [0.06, 0.15]

Analysis 1.13

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 13: Specific adverse event: psychiatric disorders

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 13: Specific adverse event: psychiatric disorders

1.13.1 Central pain ‐ multiple sclerosis

3

453

Risk Difference (IV, Random, 95% CI)

0.10 [0.05, 0.16]

1.13.2 Central pain ‐ spinal cord injury

1

116

Risk Difference (IV, Random, 95% CI)

0.00 [‐0.06, 0.07]

1.13.3 Peripheral pain ‐ chemotherapy‐induced polyneuropathy

1

36

Risk Difference (IV, Random, 95% CI)

0.11 [‐0.06, 0.28]

1.13.4 Peripheral pain ‐ diabetic polyneuropathy

1

297

Risk Difference (IV, Random, 95% CI)

0.05 [0.01, 0.09]

1.13.5 Peripheral pain ‐ polyneuropathy of various aetiologies

2

342

Risk Difference (IV, Random, 95% CI)

0.21 [0.14, 0.29]

1.13.6 Central and peripheral pain ‐ various aetiologies

1

70

Risk Difference (IV, Random, 95% CI)

0.11 [‐0.05, 0.27]

Study flow diagram

Figuras y tablas -
Figure 1

Study flow diagram

'Risk of bias' graph: review authors' judgements about each risk of bias item presented as percentages across all included studies

Figuras y tablas -
Figure 2

'Risk of bias' graph: review authors' judgements about each risk of bias item presented as percentages across all included studies

'Risk of bias' summary: review authors' judgements about each risk of bias item for each included study

Figuras y tablas -
Figure 3

'Risk of bias' summary: review authors' judgements about each risk of bias item for each included study

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 1: Pain relief of 50% or greater

Figuras y tablas -
Analysis 1.1

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 1: Pain relief of 50% or greater

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 2: Patient Global Impression much or very much improved

Figuras y tablas -
Analysis 1.2

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 2: Patient Global Impression much or very much improved

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 3: Withdrawals due to adverse events

Figuras y tablas -
Analysis 1.3

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 3: Withdrawals due to adverse events

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 4: Serious adverse events

Figuras y tablas -
Analysis 1.4

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 4: Serious adverse events

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 5: Pain relief of 30% or greater

Figuras y tablas -
Analysis 1.5

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 5: Pain relief of 30% or greater

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 6: Mean pain intensity

Figuras y tablas -
Analysis 1.6

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 6: Mean pain intensity

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 7: Health‐related quality of life

Figuras y tablas -
Analysis 1.7

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 7: Health‐related quality of life

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 8: Sleep problems

Figuras y tablas -
Analysis 1.8

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 8: Sleep problems

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 9: Psychological distress

Figuras y tablas -
Analysis 1.9

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 9: Psychological distress

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 10: Withdrawals due to lack of efficacy

Figuras y tablas -
Analysis 1.10

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 10: Withdrawals due to lack of efficacy

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 11: Any adverse event

Figuras y tablas -
Analysis 1.11

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 11: Any adverse event

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 12: Specific adverse event: nervous system disorders

Figuras y tablas -
Analysis 1.12

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 12: Specific adverse event: nervous system disorders

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 13: Specific adverse event: psychiatric disorders

Figuras y tablas -
Analysis 1.13

Comparison 1: Cannabis‐based medicines versus placebo at final treatment, Outcome 13: Specific adverse event: psychiatric disorders

Summary of findings 1. Cannabis‐based medicines compared with placebo for chronic neuropathic pain

Cannabis‐based medicines compared with placebo for chronic neuropathic pain

Patient or population: adults with chronic neuropathic pain

Settings: outpatient study centres and hospitals in Europe and North America

Intervention: cannabis‐based medicines (smoked cannabis; oral plant‐based (dronabinol) or synthetic tetrahydrocannabinol (THC) (nabilone); oromucosal spray of THC and cannabidiol (CBD))

Comparison: placebo

Outcomes

Probable outcome with intervention

95% CI

Probable outcome with placebo

Relative effect

Risk difference

(95% CI)

No. of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Participant‐reported pain relief of 50% or greater

209 per 1000

(196 to 222)

173 per 1000

0.05 (0.00 to 0.09)

1001 (8 studies)

⊕⊕⊝⊝

low1,2

NNTB 20 (11 to 100)

Patient Global Impression of Change much or very much improved

261 per 1000

(246 to 276)

211 per 1000

0.09 (0.01 to 0.17)

1092 (6 studies)

⊕⊝⊝⊝

very low1,3,4

NNTB 11 (6 to 100)

Withdrawals due to adverse events

104 per 1000

(99 to 107)

47 per 1000

0.04 (0.02 to 0.07)

1848 (13 studies)

⊕⊕⊕⊝

moderate1

NNTH 25 (16 to 50)

Serious adverse events

66 per 1000

(63 to 69)

52 per 1000

0.01 (‐0.01 to 0.03)

1876 (13 studies)

⊕⊕⊝⊝

low1,2

NNTH not calculated

Participant‐reported pain relief of 30% or greater

377 per 1000

(358 to 396)

304 per 1000

0.09 (0.03 to 0.15)

1586 (10 studies)

⊕⊕⊕⊝

moderate1

NNTB 11 (7 to 33)

Specific adverse events:nervous system disorder

611 per 1000

(576 to 644)

287 per 1000

0.38 (0.18 to 0.58)

1304 (9 studies)

⊕⊕⊝⊝

low1,3

NNTH 3 (2 to 6)

Specific adverse events:psychiatric disorders

165 per 1000

(156 to 174)

49 per 1000

0.10 (0.06 to 0.15)

1314 (9 studies)

⊕⊕⊝⊝

low1,3

NNTH 10 (7 to 16)

Abbreviations:
CI: Confidence interval; NNTB: number needed to treat for an additional beneficial outcome; NNTH: number needed to treat for an additional harmful outcome; RD: risk difference

GRADE Working Group grades of evidence

High quality: we are very confident that the true effect lies close to that of the estimate of the effect;

Moderate quality: we are moderately confident in the effect estimate; the true effect is likely to be close to the estimate of effect, but there is a possibility that it is substantially different;

Low quality: our confidence in the effect estimate is limited; the true effect may be substantially different from the estimate of the effect;

Very low quality: we have very little confidence in the effect estimate; the true effect is likely to be substantially different from the estimate of effect.

1 Downgraded once: indirectness. People with current or historical substance abuse, or both, and major medical diseases excluded.
2 Downgraded once: imprecision. CI included zero.
3 Downgraded once: inconsistency. I²>50%.

4 Downgraded once: Publication bias. All studies funded by the manufacturer of the drug.

Figuras y tablas -
Summary of findings 1. Cannabis‐based medicines compared with placebo for chronic neuropathic pain
Comparison 1. Cannabis‐based medicines versus placebo at final treatment

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1.1 Pain relief of 50% or greater Show forest plot

8

1001

Risk Difference (IV, Random, 95% CI)

0.05 [0.00, 0.09]

1.1.1 Central pain ‐ multiple sclerosis

4

669

Risk Difference (IV, Random, 95% CI)

0.08 [‐0.00, 0.15]

1.1.2 Peripheral pain ‐ chemotherapy‐induced polyneuropathy

1

36

Risk Difference (IV, Random, 95% CI)

0.11 [‐0.06, 0.28]

1.1.3 Peripheral pain ‐ diabetic polyneuropathy

1

30

Risk Difference (IV, Random, 95% CI)

‐0.20 [‐0.54, 0.14]

1.1.4 Peripheral pain ‐ plexus injury

1

141

Risk Difference (IV, Random, 95% CI)

0.01 [‐0.04, 0.06]

1.1.5 Peripheral pain ‐ polyneuropathy of various aetiologies

1

125

Risk Difference (IV, Random, 95% CI)

0.13 [0.00, 0.25]

1.2 Patient Global Impression much or very much improved Show forest plot

6

1092

Risk Difference (IV, Random, 95% CI)

0.09 [0.01, 0.17]

1.2.1 Central pain ‐ multiple sclerosis

2

397

Risk Difference (IV, Random, 95% CI)

0.06 [‐0.01, 0.14]

1.2.2 Central pain ‐ spinal cord injury

1

116

Risk Difference (IV, Random, 95% CI)

0.34 [0.17, 0.50]

1.2.3 Peripheral pain ‐ diabetic polyneuropathy

1

281

Risk Difference (IV, Random, 95% CI)

0.02 [‐0.09, 0.14]

1.2.4 Peripheral pain ‐ polyneuropathy of various aetiologies

1

228

Risk Difference (IV, Random, 95% CI)

0.08 [‐0.02, 0.17]

1.2.5 Central or peripheral pain ‐ various aetiologies

1

70

Risk Difference (IV, Random, 95% CI)

‐0.01 [‐0.22, 0.19]

1.3 Withdrawals due to adverse events Show forest plot

13

1848

Risk Difference (IV, Random, 95% CI)

0.04 [0.02, 0.07]

1.3.1 Central pain ‐ multiple sclerosis

4

693

Risk Difference (IV, Random, 95% CI)

0.04 [0.01, 0.08]

1.3.2 Central pain ‐ spinal cord injury

1

116

Risk Difference (IV, Random, 95% CI)

0.09 [0.01, 0.17]

1.3.3 Peripheral pain ‐ chemotherapy‐induced polyneuropathy

1

36

Risk Difference (IV, Random, 95% CI)

0.00 [‐0.10, 0.10]

1.3.4 Peripheral pain ‐ diabetic polyneuropathy

1

297

Risk Difference (IV, Random, 95% CI)

0.12 [0.04, 0.20]

1.3.5 Peripheral pain ‐ HIV polyneuropathy

1

68

Risk Difference (IV, Random, 95% CI)

0.00 [‐0.13, 0.13]

1.3.6 Peripheral pain ‐ plexus injury

1

141

Risk Difference (IV, Random, 95% CI)

0.01 [‐0.04, 0.06]

1.3.7 Peripheral pain ‐ polyneuropathy of various aetiologies

3

427

Risk Difference (IV, Random, 95% CI)

0.08 [0.02, 0.13]

1.3.8 Central and peripheral pain ‐ various aetiologies

1

70

Risk Difference (IV, Random, 95% CI)

‐0.06 [‐0.19, 0.07]

1.4 Serious adverse events Show forest plot

13

1876

Risk Difference (IV, Random, 95% CI)

0.01 [‐0.01, 0.03]

1.4.1 Central pain ‐ multiple sclerosis

4

693

Risk Difference (IV, Random, 95% CI)

0.03 [‐0.01, 0.06]

1.4.2 Central pain ‐ spinal cord injury

1

116

Risk Difference (IV, Random, 95% CI)

0.02 [‐0.05, 0.09]

1.4.3 Peripheral pain ‐ chemotherapy‐induced neuropathy

1

36

Risk Difference (IV, Random, 95% CI)

0.00 [‐0.10, 0.10]

1.4.4 Peripheral pain ‐ diabetic polyneuropathy

1

297

Risk Difference (IV, Random, 95% CI)

0.01 [‐0.05, 0.08]

1.4.5 Peripheral pain ‐ HIV polyneuropathy

1

68

Risk Difference (IV, Random, 95% CI)

0.03 [‐0.07, 0.13]

1.4.6 Peripheral pain ‐ plexus injury

1

141

Risk Difference (IV, Random, 95% CI)

0.00 [‐0.03, 0.03]

1.4.7 Peripheral pain ‐ polyneuropathies of various aetiologies

3

455

Risk Difference (IV, Random, 95% CI)

0.01 [‐0.02, 0.04]

1.4.8 Central and peripheral pain ‐ various aetiologies

1

70

Risk Difference (IV, Random, 95% CI)

‐0.06 [‐0.15, 0.03]

1.5 Pain relief of 30% or greater Show forest plot

10

1586

Risk Difference (IV, Random, 95% CI)

0.09 [0.03, 0.15]

1.5.1 Central pain ‐ multiple sclerosis

3

645

Risk Difference (IV, Random, 95% CI)

0.11 [‐0.03, 0.25]

1.5.2 Peripheral pain ‐ chemotherapy‐induced polyneuropathy

1

36

Risk Difference (IV, Random, 95% CI)

0.11 [‐0.16, 0.38]

1.5.3 Peripheral pain ‐ diabetic polyneuropathy

2

327

Risk Difference (IV, Random, 95% CI)

‐0.04 [‐0.14, 0.07]

1.5.4 Peripheral pain ‐ HIV polyneuropathy

1

56

Risk Difference (IV, Random, 95% CI)

0.29 [0.05, 0.52]

1.5.5 Peripheral pain ‐ plexus injury

1

141

Risk Difference (IV, Random, 95% CI)

0.10 [‐0.06, 0.25]

1.5.6 Peripheral pain ‐ polyneuropathy of various aetiologies

2

381

Risk Difference (IV, Random, 95% CI)

0.11 [0.03, 0.19]

1.6 Mean pain intensity Show forest plot

14

1837

Std. Mean Difference (IV, Random, 95% CI)

‐0.35 [‐0.60, ‐0.09]

1.6.1 Central pain ‐ multiple sclerosis

4

668

Std. Mean Difference (IV, Random, 95% CI)

‐0.10 [‐0.25, 0.05]

1.6.2 Central pain ‐ spinal cord injury

1

114

Std. Mean Difference (IV, Random, 95% CI)

‐0.04 [‐0.41, 0.33]

1.6.3 Peripheral pain ‐ chemotherapy‐induced polyneuropathy

1

36

Std. Mean Difference (IV, Random, 95% CI)

‐0.20 [‐0.86, 0.45]

1.6.4 Peripheral pain ‐ diabetic polyneuropathy

2

324

Std. Mean Difference (IV, Random, 95% CI)

‐0.05 [‐0.27, 0.17]

1.6.5 Peripheral pain ‐ HIV polyneuropathy

1

56

Std. Mean Difference (IV, Random, 95% CI)

‐0.41 [‐0.94, 0.12]

1.6.6 Peripheral pain ‐ plexus injury

1

141

Std. Mean Difference (IV, Random, 95% CI)

‐0.43 [‐0.79, ‐0.08]

1.6.7 Peripheral pain ‐ polyneuropathy of various aetiologies

3

428

Std. Mean Difference (IV, Random, 95% CI)

‐0.65 [‐1.75, 0.44]

1.6.8 Central and peripheral pain ‐ various aetiologies

1

70

Std. Mean Difference (IV, Random, 95% CI)

‐0.24 [‐0.71, 0.23]

1.7 Health‐related quality of life Show forest plot

9

1284

Std. Mean Difference (IV, Random, 95% CI)

0.02 [‐0.10, 0.13]

1.7.1 Central pain ‐ multiple sclerosis

2

363

Std. Mean Difference (IV, Random, 95% CI)

‐0.07 [‐0.27, 0.14]

1.7.2 Central pain ‐ spinal cord injury

1

113

Std. Mean Difference (IV, Random, 95% CI)

0.00 [‐0.37, 0.37]

1.7.3 Peripheral pain ‐ diabetic polyneuropathy

2

303

Std. Mean Difference (IV, Random, 95% CI)

0.17 [‐0.06, 0.39]

1.7.4 Peripheral pain ‐ plexus injury

1

141

Std. Mean Difference (IV, Random, 95% CI)

‐0.07 [‐0.42, 0.28]

1.7.5 Peripheral pain of various aetiologies

2

300

Std. Mean Difference (IV, Random, 95% CI)

‐0.03 [‐0.26, 0.21]

1.7.6 Central and peripheral pain ‐ various aetiologies

1

64

Std. Mean Difference (IV, Random, 95% CI)

0.15 [‐0.35, 0.64]

1.8 Sleep problems Show forest plot

8

1386

Std. Mean Difference (IV, Random, 95% CI)

‐0.47 [‐0.90, ‐0.04]

1.8.1 Central pain ‐ multiple sclerosis

1

339

Std. Mean Difference (IV, Random, 95% CI)

0.01 [‐0.21, 0.22]

1.8.2 Central pain ‐ spinal cord injury

1

114

Std. Mean Difference (IV, Random, 95% CI)

‐0.04 [‐0.41, 0.32]

1.8.3 Peripheral pain ‐ diabetic polyneuropathy

1

274

Std. Mean Difference (IV, Random, 95% CI)

‐0.14 [‐0.38, 0.10]

1.8.4 Peripheral pain ‐ plexus injury

1

141

Std. Mean Difference (IV, Random, 95% CI)

‐0.42 [‐0.78, ‐0.07]

1.8.5 Peripheral pain ‐ polyneuropathy of various aetiologies

3

448

Std. Mean Difference (IV, Random, 95% CI)

‐0.78 [‐2.17, 0.61]

1.8.6 Central and peripheral pain ‐ various aetiologies

1

70

Std. Mean Difference (IV, Random, 95% CI)

‐0.31 [‐0.78, 0.16]

1.9 Psychological distress Show forest plot

7

779

Std. Mean Difference (IV, Random, 95% CI)

‐0.32 [‐0.61, ‐0.02]

1.9.1 Central pain ‐ multiple sclerosis

2

363

Std. Mean Difference (IV, Random, 95% CI)

‐0.03 [‐0.65, 0.59]

1.9.2 Peripheral pain ‐ chemotherapy‐induced polyneuropathy

1

36

Std. Mean Difference (IV, Random, 95% CI)

‐1.07 [‐1.78, ‐0.37]

1.9.3 Peripheral pain ‐ diabetic polyneuropathy

1

30

Std. Mean Difference (IV, Random, 95% CI)

‐0.25 [‐0.97, 0.47]

1.9.4 Peripheral pain ‐ plexus injury

1

141

Std. Mean Difference (IV, Random, 95% CI)

‐0.27 [‐0.62, 0.08]

1.9.5 Peripheral pain ‐ polyneuropathy of various aetiologies

2

209

Std. Mean Difference (IV, Random, 95% CI)

‐0.48 [‐0.80, ‐0.16]

1.10 Withdrawals due to lack of efficacy Show forest plot

9

1576

Risk Difference (IV, Random, 95% CI)

‐0.00 [‐0.02, 0.01]

1.10.1 Central pain ‐ multiple sclerosis

4

697

Risk Difference (IV, Random, 95% CI)

0.00 [‐0.02, 0.02]

1.10.2 Peripheral pain ‐ diabetic polyneuropathy

1

297

Risk Difference (IV, Random, 95% CI)

‐0.01 [‐0.05, 0.03]

1.10.3 Peripheral pain ‐ plexus injury

1

141

Risk Difference (IV, Random, 95% CI)

0.00 [‐0.04, 0.04]

1.10.4 Peripheral pain ‐ polyneuropathy of various aetiologies

2

371

Risk Difference (IV, Random, 95% CI)

‐0.04 [‐0.09, 0.01]

1.10.5 Central and peripheral pain ‐ various aetiologies

1

70

Risk Difference (IV, Random, 95% CI)

0.00 [‐0.05, 0.05]

1.11 Any adverse event Show forest plot

7

1356

Risk Difference (IV, Random, 95% CI)

0.19 [0.12, 0.27]

1.11.1 Central pain ‐ multiple sclerosis

3

627

Risk Difference (IV, Random, 95% CI)

0.22 [0.05, 0.39]

1.11.2 Central pain ‐ spinal cord injury

1

116

Risk Difference (IV, Random, 95% CI)

0.34 [0.18, 0.50]

1.11.3 Peripheral pain ‐ diabetic polyneuropathy

1

297

Risk Difference (IV, Random, 95% CI)

0.12 [0.02, 0.22]

1.11.4 Peripheral pain ‐ polyneuropathy of various aetiologies

1

246

Risk Difference (IV, Random, 95% CI)

0.15 [0.05, 0.25]

1.11.5 Central and peripheral pain ‐ various aetiologies

1

70

Risk Difference (IV, Random, 95% CI)

0.21 [0.06, 0.36]

1.12 Specific adverse event: nervous system disorders Show forest plot

9

1304

Risk Difference (IV, Random, 95% CI)

0.38 [0.18, 0.58]

1.12.1 Central pain ‐ multiple sclerosis

3

453

Risk Difference (IV, Random, 95% CI)

0.33 [0.09, 0.58]

1.12.2 Central pain ‐ spinal cord injury

1

116

Risk Difference (IV, Random, 95% CI)

0.53 [0.38, 0.68]

1.12.3 Peripheral pain ‐chemotherapy‐induced polyneuropathy

1

36

Risk Difference (IV, Random, 95% CI)

1.00 [0.90, 1.10]

1.12.4 Peripheral pain ‐ diabetic polyneuropathy

1

297

Risk Difference (IV, Random, 95% CI)

0.26 [0.15, 0.37]

1.12.5 Peripheral pain ‐ polyneuropathy of various aetiologies

2

332

Risk Difference (IV, Random, 95% CI)

0.29 [0.19, 0.39]

1.12.6 Central and peripheral pain ‐ various aetiologies

1

70

Risk Difference (IV, Random, 95% CI)

0.37 [0.15, 0.58]

1.13 Specific adverse event: psychiatric disorders Show forest plot

9

1314

Risk Difference (IV, Random, 95% CI)

0.10 [0.06, 0.15]

1.13.1 Central pain ‐ multiple sclerosis

3

453

Risk Difference (IV, Random, 95% CI)

0.10 [0.05, 0.16]

1.13.2 Central pain ‐ spinal cord injury

1

116

Risk Difference (IV, Random, 95% CI)

0.00 [‐0.06, 0.07]

1.13.3 Peripheral pain ‐ chemotherapy‐induced polyneuropathy

1

36

Risk Difference (IV, Random, 95% CI)

0.11 [‐0.06, 0.28]

1.13.4 Peripheral pain ‐ diabetic polyneuropathy

1

297

Risk Difference (IV, Random, 95% CI)

0.05 [0.01, 0.09]

1.13.5 Peripheral pain ‐ polyneuropathy of various aetiologies

2

342

Risk Difference (IV, Random, 95% CI)

0.21 [0.14, 0.29]

1.13.6 Central and peripheral pain ‐ various aetiologies

1

70

Risk Difference (IV, Random, 95% CI)

0.11 [‐0.05, 0.27]

Figuras y tablas -
Comparison 1. Cannabis‐based medicines versus placebo at final treatment