Skip to main content

Wirksamkeit, Verträglichkeit und Sicherheit von medizinischem Cannabis

  • Chapter
Cannabis: Potenzial und Risiko

Zusammenfassung

Viele Erkrankungen können von konstanten oder intermittierenden, anhaltenden Schmerzen begleitet werden. Dies ist beispielsweise häufig der Fall bei Krebserkrankungen, multipler Sklerose (MS) (Abschn. 4.2 und Abschn. 4.4), entzündlich rheumatischen Erkrankungen oder neuropathischen Schmerzen (aufgrund von HIV-Infektionen, Herpes zoster, Diabetes mellitus, Zytostatikabehandlung, Rückenmarkverletzungen, Operationen etc.).

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 59.99
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 74.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Preview

Unable to display preview. Download preview PDF.

Unable to display preview. Download preview PDF.

Literatur

  1. Abrams DI, Jay CA, Shade SB, Vizoso H, Reda H, Press S (2007) Cannabis in painful HIV-associated sensory neuropathy: a randomized placebo-controlled trial. Neurology 68 (7): 515–521

    Google Scholar 

  2. Andreae MH, Carter GM, Shaparin N, Suslov K, Ellis RJ, Ware MA, Abrams DI, Prasad H, Wilsey B, Indyk D, Johnson M, Sacks HS (2015) Inhaled cannabis for chronic neuropathic pain: a meta-analysis of individual patient data. J Pain 16 (12): 1221–1232. doi: 10.1016/j.jpain.2015.07.009

  3. Berman JS, Symonds C, Birch R (2004) Efficacy of two cannabis based medicinal extracts for relief of central neuropathic pain from brachial plexus avulsion: results of a randomised controlled trial. Pain 112: 299–306

    Google Scholar 

  4. Blake DR, Robson P, Ho M, Jubb RW, McCabe CS (2006) Preliminary assessment of the efficacy, tolerability and safety of a cannabis-based medicine (Sativex) in the treatment of pain caused by rheumatoid arthritis. Rheumatology (Oxford) 45 (1): 50–52

    Google Scholar 

  5. Bonica JJ (2001) Bonica’s management of pain. Lippincott Williams & Wilkins, Philadelphia

    Google Scholar 

  6. Boychuk DG, Goddard G, Mauro G, Orellana MF (2015) The effectiveness of cannabinoids in the management of chronic nonmalignant neuropathic pain: a systematic review. J Oral Facial Pain Headache 29 (1): 7–14. doi: 10.11607/ofph.1274

  7. Corey-Bloom J, Wolfson T, Gamst A, Jin S, Marcotte TD, Bentley H, et al. (2012) Smoked cannabis for spasticity in multiple sclerosis: a randomized, placebo controlled trial. Can Med Assoc J 184 (10): 1143–1150

    Google Scholar 

  8. de Vries M, van Rijckevorsel DCM, Wilder-Smith OHG, van Goor H (2014) Dronabinol and chronic pain: importance of mechanistic considerations. Expert Op Pharmacother 15 (11): 1525–1534. doi: 10.1517/14656566.2014.918102

  9. Dempster B (2003) Clinical study report: a multi centre randomised, double blind, placebo controlled, parallel group comparison of the effects of cannabis based medicine standardised extracts over 4 weeks, in patients with chronic refractory pain dure to multiple sclerosis or other defects of neurological function. [Study code: GMNS0107, 1–237. GW Pharma Ltd].

    Google Scholar 

  10. Deshpande A, Mailis-Gagnon A, Zoheiry N, Lakha SF (2015) Efficacy and adverse effects of medical marijuana for chronic noncancer pain: Systematic review of randomized controlled trials. Can Family Physician 61: e372–381

    Google Scholar 

  11. Dworkin RH, Turk DC, Farrar JT, Haythornthwaite JA, Jensen MP, Katz NP, Kerns RD, Stucki G, Allen RR, Bellamy N, Carr DB, Chandler J, Cowan P, Dionne R, Galer BS, Hertz S, Jadad AR, Kramer LD, Manning DC, Martin S, McCormick CG, McDermott MP, McGrath P, Quessy S, Rappaport BA, Robbins W, Robinson JP, Rothman M, Royal MA, Simon L, Stauffer JW, Stein W, Tollett J, Wernicke J, Witter J (2005) Core outcome measures for chronic pain clinical trials: IMMPACT recommendations. Pain 113 (1–2): 9–19. doi: 10.1016/j.pain.2004.09.012

  12. Ellis RJ, Toperoff W, Vaida F, van den Brande G, Gonzales J, Gouaux B, et al. (2009) Smoked medicinal cannabis for neuropathic pain in HIV: a randomized, crossover clinical trial. Neuropsychopharmacology 34 (3): 672–680.

    Google Scholar 

  13. Finnerup NB, Attal N, Haroutounian S, McNicol E, Baron R, Dworkin RH, Gilron I, Haanpaa M, Hansson P, Jensen TS, Kamerman PR, Lund K, Moore A, Raja SN, Rice AS, Rowbotham M, Sena E, Siddall P, Smith BH, Wallace M (2015) Pharmacotherapy for neuropathic pain in adults: a systematic review and meta-analysis. Lancet Neurol 14 (2): 162–173. doi: 10.1016/s1474-4422(14)70251-0

  14. Fitzcharles MA, Baerwald C, Ablin J, Häuser W (2016) Efficacy, tolerability and safety of cannabinoids in chronic pain associated with rheumatic diseases (fibromyalgia syndrome, back pain, osteoarthritis, rheumatoid arthritis). Schmerz 30: 47–61

    Google Scholar 

  15. Guindon J, Hohmann AG (2009) The endocannabinoid system and pain. CNS Neurol Disorders Drug Targets (8): 403–429.

    Google Scholar 

  16. GW Pharmaceuticals Ltd. [Clinical-Trials.gov identifier NCT00391079]. US National Institutes of Health, ClinicalTrials.gov.

    Google Scholar 

  17. Hillard CJ, Weinlander KM, Stuhr KL (2012) Contributions of endocannabinoid signaling to psychiatric disorders in humans: genetic and biochemical evidence. Neuroscience (204): 207–229

    Google Scholar 

  18. Iskedjian M, Bereza B, Gordon A, Piwko C, Einarson TR (2007) Meta-analysis of cannabis based treatments for neuropathic and multiple sclerosis-related pain. Curr Med Res Opinion 23 (1): 17–24. doi: http://dx.doi.org/10.1185/030079906X158066

  19. Jadad AR, Moore RA, Carroll D, Jenkinson C, Reynolds DJ, Gavaghan DJ, al. e (1996) Assessing the quality of reports of randomized clinical trials: is blinding necessary? Controlled Clin Trials 17 (1): 1–12

    Google Scholar 

  20. Jawahar R, Oh U, Yang S, Lapane KL (2013) A systematic review of pharmacological pain management in multiple sclerosis. Drugs 73: 1711–1722

    Google Scholar 

  21. Ji RR, Chamessian A, Zhang YQ (2016) Pain regulation by non-neuronal cells and inflammation. Science 254 (6312): 572–577

    Google Scholar 

  22. Johnson JR, Burnell-Nugent M, Lossignol D, Ganae-Motan ED, Potts R, Fallon MT (2010) Multicenter, double-blind, randomized, placebo-controlled, parallel-group study of the efficacy, safety, and tolerability of THC: CBD extract and THC extract in patients with intractable cancer-related pain. J Pain Symptom Manag 2 (39): 167–179

    Google Scholar 

  23. Karst M, Salim K, Burstein S, al. e (2003) Analgetic effect of the synthetic cannabinoid CT-3 on chronic neuropathic pain: a randomized controlled trial. J Am Med Assoc 290: 1757–1762

    Google Scholar 

  24. Lewin S, Glenton C, Munthe-Kaas H, Carlsen B, Colvin CJ, Gulmezoglu M, Noyes J, Booth A, Garside R, Rashidian A (2015) Using qualitative evidence in decision making for health and social interventions: an approach to assess confidence in findings from qualitative evidence syntheses (GRADE-CERQual). PLoS Med 12 (10): e1001895. doi: 10.1371/journal.pmed.1001895

  25. Lynch ME, Cesar-Rittenberg P, Hohmann AG (2014) A double-blind, placebo-controlled, corssover pilot trial with extension using oral mucosal cannabinoid extract for treatment of chemotherapy-induced neuropathic pain. J Pain Symptom Manag 47: 166–173 23742737

    Google Scholar 

  26. Martin-Sanchez E, Furukawa TA, Taylor J, Martin JL (2009) Systematic review and meta-analysis of cannabis treatment for chronic pain. Pain Med 10 (8): 1353–1368. doi: http://dx.doi.org/10.1111/j.1526-4637.2009.00703.x

  27. McQuay HJ, Kalso E, Moore RA (eds) (2008) Systematic reviews in pain research: methodology refined. IASP Press, Seattle

    Google Scholar 

  28. Melzack R (1975) The McGill Pain Questionnaire: Major properties and scoring methods. Pain 1 (3): 277–299

    Google Scholar 

  29. Merskey H, Bogduk N (2013) Classification of chronic pain: descriptions of chronic pain syndromes and definitions of pain terms. 2013. International Association for the Study of Pain (IASP), Task Force on Taxonomy, Seattle 1994. International Association for the Study of Pain (IASP), Task Force on Taxonomy, Seattle

    Google Scholar 

  30. Mücke M, Carter C, Cuhls H, Prüß M, Radbruch L, Häuser W (2016) Cannabinoide in der palliativen Versorgung – Systematische Übersicht und Metaanalyse der Wirksamkeit, Verträglichkeit und Sicherheit. Schmerz 30: 25–36

    Google Scholar 

  31. Noyes RJ, Brunk SF, AVery DA, Canter A (1975a) The analgesic properties of the delta-9-tetrahydrocannabinol and codeine. Clin Pharmacol Ther 18 (1): 84–89

    Google Scholar 

  32. Noyes RJ, Brunk SF, Baram DA, Canter A (1975b) Analgesic effect of delta-9-tetrahydrocannabinol. J Clinical Pharmacology 15 (2–3): 139–143

    Google Scholar 

  33. OCEBM (2011) The Oxford Levels of Evidence 2. Levels of Evicence Working Group. http://www.cebm.net/index.aspx?o = 5653, 2 edn. Oxford Centre for Evidence-Based Medicine, Oxford

  34. Owens B (2015) Drug development: The treasure chest. Nature (525): S6-S8.

    Google Scholar 

  35. Petzke F, Enax-Krumova EK, Häuser W (2016) Wirksamkeit, Verträglichkeit und Sicherheit von Cannabinoiden bei neuropathischen Schmerzsyndromen – Eine systematische Übersichtsarbeit von randomisierten, kontrollierten Studien. Schmerz 30: 62–88

    Google Scholar 

  36. Pinsger M, Schimetta W, Volc D, Hiermann E, Riederer F, Pölz W (2006) Benefits of an add-on treatment with the synthetic cannabinomimetic nabilone on patients with chronic pain – a randomized controlled trial. Wiener Klin Wchenschr 118 (11–12): 327–335

    Google Scholar 

  37. Portenoy RK, Ganae-Motan ED, Allende S, Yanagihara R, Shaiova L, Weinstein S, McQuade R, Wright S, Fallon MT (2012) Nabiximols for opioid-treated cancer patients with poorly-controlled chronic pain: a randomized, placebo-controlled, graded-dose trial. J Pain 13 (5): 438–449

    Google Scholar 

  38. Rog DJ, Nurmikko TJ, Friede T, Young CA (2005) Randomized, controlled trial of cannabis-based medicine in central pain in multiple sclerosis. Neurology 65: 812–819

    Google Scholar 

  39. Russo EB (2008) Clinical endocannabinoid deficiency (CECD): can this concept explain therapeutic benefits of cannabis in migraine, fibromyalgia, irritable bowel syndrome and other treatment-resistant conditions? Neuroendocrinol Lett (29): 192–200

    Google Scholar 

  40. Serpell M, Ratcliffe S, Hovorka J, Schofield M, Taylor L, Lauder H, Ehler E (2014) A double-blind, randomized, placebo-controlled, parallel group study of THC/CBD spray in peripheral neuropathic pain treatment. Eur J Pain 18 (7): 999–1012. doi: 10.1002/j.1532-2149.2013.00445.x

  41. Skrabek RQ, Galimova L, Ethans K, Perry D (2008) Nabilone for the treatment of pain in fibromyalgia. J Pain 9 (2): 164–173

    Google Scholar 

  42. Staquet M, Gantt C, Machin D (1978) Effect of a nitrogen analog oftetrahydrocannabinol on cancer pain. Clin Pharmacol Ther 23 (4): 397–401

    Google Scholar 

  43. Svendsen KB, Jensen TS, Bach FW (2004) Does the cannabinoid dronabinol reduce central pain in multiple sclerosis? Randomised double blind placebo controlled crossover trial. Br Med J (online) 329 (7460): 253

    Google Scholar 

  44. Thomasius R, Petersen KU (2007) Auwirkungen von Cannabiskonsum und -missbrauch. Eine Expertise zu gesundheitlichen und psychosozialen Folgen. 1 systematisches Review der international publizierten Studien von 1996–2006. Pabst Science Publishers, Lengerich

    Google Scholar 

  45. Van Houdenhove B, Egle UT (2004) Fibromyalgia: a stress disorder? Piecing the biopsychosocial puzzle together. Psychother Psychosomat (73): 267–275

    Google Scholar 

  46. van Riel PL, Fransen J (2005) DAS28: a useful instrument to monitor infliximab treatment in patients with rheumatoid arthritis. Arthritis research & therapy 7 (5): 189–190. doi: 10.1186/ar1820

  47. Vardeh D, Mannion RJ, Woolf C (2016) Toward a mechanism based approach to pain diagnosis. J Pain 17 (9): T50–69

    Google Scholar 

  48. Wade DT, Makela P, Robson P, House H, Bateman C (2004) Do cannabis-based medicinal extracts have general or specific effects on symptoms in multiple sclerosis? A double-blind, randomized, placebo-controlled study on 160 patients. Multiple Sclerosis 10 (4): 434–441

    Google Scholar 

  49. Wade DT, Robson P, House H, Makela P, Aram J (2003) A preliminary controlled study to determine whether whole-plant cannabis extracts can improve intractable neurogenic symptoms. Clin Rehabil 17 (1): 21–29

    Google Scholar 

  50. Walitt B, Klose P, Fitzcharles MA, Phillips T, Häuser W (2016) Cannabinoids for fibromyalgia. Cochrane Database Syst Rev (7). doi: 10.1002/14651858.CD011694.pub2

  51. Ware JE, Maruish MEE (1999) SF-36 Health Survey. The use of psychological testing for treatment planning and outcomes assessment, 2nd ed. Lawrence Erlbaum Associates Publishers, Mahwah, NJ, US, xvi, pp 1227–1246 (1507 pp)

    Google Scholar 

  52. Ware MA, Wang T, Shapiro S, Robinson A, Ducruet T, Huynh T, al. e (2010) Smoked cannabis for chronic neuropathic pain: a randomized controlled trial. Can Med Assoc J 182 (14): E694–701

    Google Scholar 

  53. Whiting P, Savovic J, Higgins JP, Caldwell DM, Reeves BC, Shea B, Davies P, Kleijnen J, Churchill R (2016) ROBIS: A new tool to assess risk of bias in systematic reviews was developed. J Clin Epidemiol 69: 225–234. doi: 10.1016/j.jclinepi.2015.06.005

  54. Whiting PF, Wolff RF, Deshpande S, Di Nisio M, Duffy S, Hernandez AV, Keurentjes JC, Lang S, Misso K, Ryder S, Schmidlkofer S, Westwood M, Kleijnen J (2015) Cannabinoids for medical use: A systematic review and meta-analysis. J Am Med Assoc 313 (24): 2456–2473. doi: 10.1001/jama.2015.6358

  55. Wiech K (2016) Deconstructing the sensation of pain: the influence of cognitive processes on pain perception. Science 354 (6312): 584–587

    Google Scholar 

  56. Wilsey B, Marcotte T, Deutsch R, Gouaux B, Sakai S, Donaghe H (2013) Low dose vaporized cannabis significantly improves neuropathic pain. J Pain 14 (2): 136–148

    Google Scholar 

  57. Wilsey B, Marcotte T, Tsodikov A, Millman J, Bentley H, Gouaux B, al. e (2008) A randomized, placebo-controlled, crossover trial of cannabis cigarettes in neuropathic pain. J Pain 9 (6): 506–521

    Google Scholar 

  58. Wissel J, Haydn T, Muller J, al. e (2006) Low dose treatment with the synthetic cannabinoid Nabilone significantly reduces spasticity-related pain: A double-blind placebo-controlled cross-over trial. J Neurol 253 (10): 1337–1341

    Google Scholar 

  59. Xanthos DN, Sandkühler J (2014) Neurogenic neuroinflammation: inflammatory CNS reactions in response to neuronal activity. Nat Rev Neurosci 15 (1): 43–53

    Google Scholar 

  60. Zhang J, Echeverry S, Lim TK, Lee SH, Shi XQ, Huang H (2015) Can modulating inflammatory response be a good strategy to treat neuropathic pain? Curr Pharmaceut Design (21): 831–839

    Google Scholar 

  61. Barnes M, Kent R, Semlyen J, McMullen K (2003) Spasticity in multiple sclerosis. Neurorehabil Neural Repair 17 (1): 66–70

    Google Scholar 

  62. Bohannon R, Smith M (1987) Interrater reliability of a modified Ashworth scale of muscle spasticity. Phys Ter 67 (2): 206–207

    Google Scholar 

  63. Centonze D, Mori F, Koch G, Buttari F, Codecà C, Rossi S, Cencioni MT, Bari M, Fiore S, Bernardi G (2009) Lack of effect of cannabis-based treatment on clinical and laboratory measures in multiple sclerosis. Neurol Sci 30 (6): 531

    Google Scholar 

  64. Collin C, Davies P, Mutiboko I, Ratcliffe S (2007) Randomized controlled trial of cannabis-based medicine in spasticity caused by multiple sclerosis. Eur J Neurol 14 (3): 290–296

    Google Scholar 

  65. Collin C, Ehler E, Waberzinek G, Alsindi Z, Davies P, Powell K, Notcutt W, O’leary C, Ratcliffe S, Nováková I (2010) A double-blind, randomized, placebo-controlled, parallel-group study of Sativex, in subjects with symptoms of spasticity due to multiple sclerosis. Neurol Res 32 (5): 451–459

    Google Scholar 

  66. Corey-Bloom J, Wolfson T, Gamst A, Jin S, Marcotte TD, Bentley H, Gouaux B (2012) Smoked cannabis for spasticity in multiple sclerosis: a randomized, placebo-controlled trial. Can Med Assoc J 184 (10): 1143–1150

    Google Scholar 

  67. Fife TD, Moawad H, Moschonas C, Shepard K, Hammond N (2015) Clinical perspectives on medical marijuana (cannabis) for neurologic disorders. Neurol Clin Pract 5 (4): 344–351. doi: http://dx.doi.org/10.1212/CPJ.0000000000000162

  68. Fleuren J, Voerman G, Erren-Wolters C, Snoek G, Rietman J, Hermens H, Nene A (2010) Stop Using the Ashworth Scale for the assesment of spasticity. J Neurol Neurosurg Psychiat 81 (1): 46–52

    Google Scholar 

  69. Gowran A, Noonan J, Campbell VA (2011) The multiplicity of action of cannabinoids: implications for treating neurodegeneration. CNS Neurosci Ther 17 (6): 637–644

    Google Scholar 

  70. Greenberg HS, Werness SA, Pugh JE, Andrus RO, Anderson DJ, Domino EF (1994) Short-term effects of smoking marijuana on balance in patients with multiple sclerosis and normal volunteers. Clin Pharmacol Ther 55 (3): 324–328

    Google Scholar 

  71. Killestein J, Hoogervorst E, Reif M, Kalkers N, Van Loenen A, Staats P, Gorter R, Uitdehaag B, Polman C (2002) Safety, tolerability, and efficacy of orally administered cannabinoids in MS. Neurology 58 (9): 1404–1407

    Google Scholar 

  72. Koppel BS, Brust JCM, Fife T, Bronstein J, Youssof S, Gronseth G, Gloss D (2014) Systematic review: Efficacy and safety of medical marijuana in selected neurologic disorders: Report of the Guideline Development Subcommittee of the American Academy of Neurology. Neurology 82 (17): 1556–1563. doi: 10.1212/WNL.0000000000000363

  73. Kubajewska I, Constantinescu CS (2010) Cannabinoids and experimental models of multiple sclerosis. Immunobiology 215 (8): 647–657

    Google Scholar 

  74. Leussink VI, Husseini L, Warnke C, Broussalis E, Hartung H-P, Kieseier BC (2012) Symptomatic therapy in multiple sclerosis: the role of cannabinoids in treating spasticity. Ther Adv Neurol Disord 5 (5): 255–266

    Google Scholar 

  75. Lewin S, Glenton C, Munthe-Kaas H, Carlsen B, Colvin CJ, Gulmezoglu M, Noyes J, Booth A, Garside R, Rashidian A (2015) Using qualitative evidence in decision making for health and social interventions: an approach to assess confidence in findings from qualitative evidence syntheses (GRADE-CERQual). PLoS Med 12 (10): e1001895. doi: 10.1371/journal.pmed.1001895

  76. OCEBM (2011) Levels of Evidence Working Group. The Oxford Levels of Evidence 2. http://www.cebm.net/index.aspx?o = 5653, 2 edn. Oxford Centre for Evidence-Based Medicine, Oxford

  77. Pryce G, Baker D (2007) Control of spasticity in a multiple sclerosis model is mediated by CB1, not CB2, cannabinoid receptors. Br J Pharmacol 150 (4): 519–525

    Google Scholar 

  78. Rog DJ, Nurmikko TJ, Friede T, Young CA (2005) Randomized, controlled trial of cannabis-based medicine in central pain in multiple sclerosis. Neurology 65 (6): 812–819

    Google Scholar 

  79. Rossi S, Bernardi G, D C (2010) The endocannabinoid system in the inflammatory and neurodegenerative processes of multiple sclerosis and of amyotrophic lateral sclerosis. Exp Neurol 224 (1): 92–102

    Google Scholar 

  80. Sanchez A, Garcia-Merino A (2012) Neuroprotective agents: cannabinoids. Clin Immunol 142 (1): 57–67

    Google Scholar 

  81. Thomasius R, Petersen KU (2007) Auwirkungen von Cannabiskonsum und -missbrauch. Eine Expertise zu gesundheitlichen und psychosozialen Folgen. 1 systematisches Review der international publizierten Studien von 1996–2006. Pabst Science Publishers, Lengerich

    Google Scholar 

  82. Ungerleider JT, Andyrsiak T, Fairbanks L, Ellison GW, Myers LW (1988) Delta-9-THC in the treatment of spasticity associated with multiple sclerosis. Adv Alcohol Subst Abuse 7 (1): 39–50

    Google Scholar 

  83. Vaney C, Heinzel-Gutenbrunner M, Jobin P, Tschopp F, Gattlen B, Hagen U, Schnelle M, Reif M (2004) Efficacy, safety and tolerability of an orally administered cannabis extract in the treatment of spasticity in patients with multiple sclerosis: a randomized, double-blind, placebo-controlled, crossover study. Multiple Sclerosis 10 (4): 417–424

    Google Scholar 

  84. Wade DT, Makela P, House H, Bateman C, Robson P (2006) Long-term use of a cannabis-based medicine in the treatment of spasticity and other symptoms in multiple sclerosis. Multiple Sclerosis J 12 (5): 639–645

    Google Scholar 

  85. Whiting P, Savovic J, Higgins JP, Caldwell DM, Reeves BC, Shea B, Davies P, Kleijnen J, Churchill R (2016) ROBIS: A new tool to assess risk of bias in systematic reviews was developed. J Clin Epidemiol 69: 225–234. doi: 10.1016/j.jclinepi.2015.06.005

  86. Whiting PF, Wolff RF, Deshpande S, Di Nisio M, Duffy S, Hernandez AV, Keurentjes JC, Lang S, Misso K, Ryder S, Schmidlkofer S, Westwood M, Kleijnen J (2015) Cannabinoids for medical use: A systematic review and meta-analysis. J Am Med Assoc 313 (24): 2456–2473. doi: 10.1001/jama.2015.6358

  87. Zajicek J, Fox P, Sanders H, Wright D, Vickery J, Nunn A, Thompson A, group UMr (2003) Cannabinoids for treatment of spasticity and other symptoms related to multiple sclerosis (CAMS study): multicentre randomised placebo-controlled trial. Lancet 362 (9395): 1517–1526

    Google Scholar 

  88. Zajicek J, Reif M, Schnelle M Cannabis extract in the treatment of muscle stiffness and other symptoms in multiple sclerosis-results of the MUSEC study. In: Multiple Sclerosis, 2009. vol 9. SAGE Publications Ltd 1 Olivers Yard, 55 City Road, London EC1Y 1SP, England, pp S274–S274

    Google Scholar 

  89. Zajicek J, Sanders H, Wright D, Vickery P, Ingram W, Reilly S, Nunn A, Teare L, Fox P, Thompson A (2005) Cannabinoids in multiple sclerosis (CAMS) study: safety and efficacy data for 12 months follow up. J Neurol Neurosurg Psychiat 76 (12): 1664–1669

    Google Scholar 

  90. Zajicek JP, Hobart JC, Slade A, Barnes D, Mattison PG, Group MR (2012) Multiple sclerosis and extract of cannabis: results of the MUSEC trial. J Neurol Neurosurg Psychiat 83 (11): 1125–1132

    Google Scholar 

  91. Abrams DI, Hilton JF, Leiser RJ, Shade SB, Elbeik TA, Aweeka FT, Benowitz NL, Bredt BM, Kosel B, Aberg JA, Deeks SG, Mitchell TF, Mulligan K, Bacchetti P, McCune JM, Schambelan M (2003) Short-term effects of cannabinoids in patients with HIV-1 infection: a randomized, placebo-controlled clinical trial. Ann Intern Med 139 (4): 258–266

    Google Scholar 

  92. Beal JE, Olson R, Laubenstein L, Morales JO, Bellman P, Yangco B, Lefkowitz L, Plasse TF, Shepard KV (1995) Dronabinol as a treatment for anorexia associated with weight loss in patients with AIDS. J Pain Sympt Manag 10 (2): 89–97

    Google Scholar 

  93. Brisbois TD, de Kock IH, Watanabe SM, Mirhosseini M, Lamoureux DC, Chasen M, MacDonald N, Baracos VE, Wismer WV (2011) Delta-9-tetrahydrocannabinol may palliate altered chemosensory perception in cancer patients: results of a randomized, double-blind, placebo-controlled pilot trial. Ann Oncol 22 (9): 2086–2093. doi: 10.1093/annonc/mdq727

  94. Chan HS, Correia JA, MacLeod SM (1987) Nabilone versus prochlorperazine for control of cancer chemotherapy-induced emesis in children: a double-blind, crossover trial. Pediatrics 79 (6): 946–952

    Google Scholar 

  95. Dupuis LL, Roscoe JA, Olver I, Aapro M, Molassiotis A (2017) 2016 updated MASCC/ESMO consensus recommendations: Anticipatory nausea and vomiting in children and adults receiving chemotherapy. Support Care Cancer 25 (1): 317–321. doi: 10.1007/s00520-016-3330-z

  96. Duran M, Perez E, Abanades S, Vidal X, Saura C, Majem M, Arriola E, Rabanal M, Pastor A, Farre M, Rams N, Laporte JR, Capella D (2010) Preliminary efficacy and safety of an oromucosal standardized cannabis extract in chemotherapy-induced nausea and vomiting. Br J Clin Pharmacol 70 (5): 656–663. doi: 10.1111/j.1365-2125.2010.03743.x

  97. Friemel CM, Zimmer A, Schneider M (2014) The CB1 receptor as an important mediator of hedonic reward processing. Neuropsychopharmacology 39 (10): 2387–2396. doi: 10.1038/npp.2014.86

  98. Guyatt G, Oxman AD, Akl EA, Kunz R, Vist G, Brozek J, Norris S, Falck-Ytter Y, Glasziou P, DeBeer H, Jaeschke R, Rind D, Meerpohl J, Dahm P, Schunemann HJ (2011) GRADE guidelines: 1. Introduction-GRADE evidence profiles and summary of findings tables. J Clin Epidemiol 64 (4): 383–394. doi: 10.1016/j.jclinepi.2010.04.026

  99. Haney M, Gunderson EW, Rabkin J, Hart CL, Vosburg SK, Comer SD, Foltin RW (2007) Dronabinol and marijuana in HIV-positive marijuana smokers. Caloric intake, mood, and sleep. J Acquir Immun Deficienc Syndroms (1999) 45 (5): 545–554. doi: 10.1097/QAI.0b013e31811ed205

  100. Haney M, Rabkin J, Gunderson E, Foltin RW (2005) Dronabinol and marijuana in HIV (+) marijuana smokers: acute effects on caloric intake and mood. Psychopharmacology 181 (1): 170–178. doi: 10.1007/s00213-005-2242-2

  101. Jatoi A, Windschitl HE, Loprinzi CL, Sloan JA, Dakhil SR, Mailliard JA, Pundaleeka S, Kardinal CG, Fitch TR, Krook JE, Novotny PJ, Christensen B (2002) Dronabinol versus megestrol acetate versus combination therapy for cancer-associated anorexia: a North Central Cancer Treatment Group study. J Society of Clin Oncol 20 (2): 567–573. doi: 10.1200/jco.2002.20.2.567

  102. Johnson JR, Burnell-Nugent M, Lossignol D, Ganae-Motan ED, Potts R, Fallon MT (2010) Multicenter, double-blind, randomized, placebo-controlled, parallel-group study of the efficacy, safety, and tolerability of THC: CBD extract and THC extract in patients with intractable cancer-related pain. J Pain Sympt Manag 39 (2): 167–179. doi: 10.1016/j.jpainsymman.2009.06.008

  103. Kamen C, Tejani MA, Chandwani K, Janelsins M, Peoples AR, Roscoe JA, Morrow GR (2014) Anticipatory nausea and vomiting due to chemotherapy. Eur J Pharmacol 0: 10.1016/j.ejphar.2013.1009.1071. doi: 10.1016/j.ejphar.2013.09.071

  104. Kirkham TC (2009) Cannabinoids and appetite: food craving and food pleasure. Int Rev Psychiat 21 (2): 163–171. doi: 10.1080/09540260902782810

  105. Koch M, Varela L, Kim JG, Kim JD, Hernandez-Nuno F, Simonds SE, Castorena CM, Vianna CR, Elmquist JK, Morozov YM, Rakic P, Bechmann I, Cowley MA, Szigeti-Buck K, Dietrich MO, Gao XB, Diano S, Horvath TL (2015) Hypothalamic POMC neurons promote cannabinoid-induced feeding. Nature 519 (7541): 45–50. doi: 10.1038/nature14260

  106. Lewin S, Glenton C, Munthe-Kaas H, Carlsen B, Colvin CJ, Gulmezoglu M, Noyes J, Booth A, Garside R, Rashidian A (2015) Using qualitative evidence in decision making for health and social interventions: an approach to assess confidence in findings from qualitative evidence syntheses (GRADE-CERQual). PLoS Med 12 (10): e1001895. doi: 10.1371/journal.pmed.1001895

  107. Lutge EE, Gray A, Siegfried N (2013) The medical use of cannabis for reducing morbidity and mortality in patients with HIV/AIDS. Cochrane Database Syst Rev 4: CD005175. doi: http://dx.doi.org/10.1002/14651858.CD005175.pub3

  108. Machado Rocha FC, Stefano SC, De Cassia Haiek R, Rosa Oliveira LMQ, Da Silveira DX (2008) Therapeutic use of Cannabis sativa on chemotherapy-induced nausea and vomiting among cancer patients: Systematic review and meta-analysis. Eur J Cancer Care 17 (5): 431–443. doi: http://dx.doi.org/10.1111/j.1365-2354.2008.00917.x

  109. Mechoulam R, Hanus L (2001) The cannabinoids: an overview. Therapeutic implications in vomiting and nausea after cancer chemotherapy, in appetite promotion, in multiple sclerosis and in neuroprotection. Pain Res Manag 6 (2): 67–73

    Google Scholar 

  110. MedizInfo Anorexie-Kachexie-Syndrom. MedizInfo (R). http://www.medizinfo.de/palliativmedizin/ernaehrung/anorexie_kachexie_syndrom.shtml. Accessed 15.07.2017

  111. Meiri E, Jhangiani H, Vredenburgh JJ, Barbato LM, Carter FJ, Yang HM, Baranowski V (2007) Efficacy of dronabinol alone and in combination with ondansetron versus ondansetron alone for delayed chemotherapy-induced nausea and vomiting. Curr Med Res Opin 23 (3): 533–543. doi: 10.1185/030079907X167525

  112. Melhem-Bertrandt AI, Munsell MF, Fisch M, al. (2014) A randomized, double-blind, placebo-controlled trial of palonosetron plus dexamethasone with or without dronabinol for the prevention of chemotherapy-induced nausea and vomiting after moderately emetogenic chemotherapy [Unpublished manuscript].

    Google Scholar 

  113. Mücke M, Carter C, Cuhls H, Pruss M, Radbruch L, Hauser W (2016) [Cannabinoids in palliative care: Systematic review and meta-analysis of efficacy, tolerability and safety]. Schmerz (Berlin, Germany) 30 (1): 25–36. doi: 10.1007/s00482-015-0085-2

  114. OCEBM (2011) The Oxford Levels of Evidence 2. Levels of Evidence Working Group. http://www.cebm.net/index.aspx?o=5653, 2 edn. Oxford Centre for Evidence-Based Medicine, Oxford

  115. Parker LA, Rock EM, Limebeer CL (2011) Regulation of nausea and vomiting by cannabinoids. Br J Pharmacol 163 (7): 1411–1422. doi: 10.1111/j.1476-5381.2010.01176.x

  116. Pertwee RG (2009) Emerging strategies for exploiting cannabinoid receptor agonists as medicines. Br J Pharmacol 156 (3): 397–411. doi: 10.1111/j.1476-5381.2008.00048.x

  117. Phillips RS, Friend AJ, Gibson F, Houghton E, Gopaul S, Craig JV, Pizer B (2016) Antiemetic medication for prevention and treatment of chemotherapy-induced nausea and vomiting in childhood. Cochrane Database Syst Rev 2016 (2) (no pagination) (CD007786). doi: http://dx.doi.org/10.1002/14651858.CD007786.pub3

  118. Rock EM, Parker LA (2016) Cannabinoids as potential treatment for chemotherapy-induced nausea and vomiting. Front Pharmacol 7: 221. doi: 10.3389/fphar.2016.00221

  119. Smith LA, Azariah F, Lavender VT, Stoner NS, Bettiol S (2015) Cannabinoids for nausea and vomiting in adults with cancer receiving chemotherapy. Cochrane Database Syst Rev (11): Cd009464. doi: 10.1002/14651858.CD009464.pub2

  120. Steinman J, DeBoer MD (2013) Treatment of cachexia: melanocortin and ghrelin interventions. Vitamins Hormones 92: 197–242. doi: 10.1016/b978-0-12-410473-0.00008-8

  121. Strasser F, Luftner D, Possinger K, Ernst G, Ruhstaller T, Meissner W, Ko YD, Schnelle M, Reif M, Cerny T (2006) Comparison of orally administered cannabis extract and delta-9-tetrahydrocannabinol in treating patients with cancer-related anorexia-cachexia syndrome: a multicenter, phase III, randomized, double-blind, placebo-controlled clinical trial from the Cannabis-In-Cachexia-Study-Group. J Society of Clin Oncol 24 (21): 3394–3400. doi: 10.1200/jco.2005.05.1847

  122. Struwe M, Kaempfer SH, Geiger CJ, Pavia AT, Plasse TF, Shepard KV, Ries K, Evans TG (1993) Effect of dronabinol on nutritional status in HIV infection. Ann Pharmacother 27 (7–8): 827–831. doi: 10.1177/106002809302700701

  123. Tafelski S, Hauser W, Schafer M (2016) Efficacy, tolerability, and safety of cannabinoids for chemotherapy-induced nausea and vomiting--a systematic review of systematic reviews. Schmerz (Berlin, Germany) 30 (1): 14–24. doi: 10.1007/s00482-015-0092-3

  124. Thomasius R, Petersen KU (2007) Auwirkungen von Cannabiskonsum und -missbrauch. Eine Expertise zu gesundheitlichen und psychosozialen Folgen. 1 systematisches Review der international publizierten Studien von 1996–2006. Pabst Science Publishers, Lengerich

    Google Scholar 

  125. Timpone JG, Wright DJ, Li N, Egorin MJ, Enama ME, Mayers J, Galetto G (1997) The safety and pharmacokinetics of single-agent and combination therapy with megestrol acetate and dronabinol for the treatment of HIV wasting syndrome. The DATRI 004 Study Group. Division of AIDS Treatment Research Initiative. AIDS Res Human Retrovir 13 (4): 305–315. doi: 10.1089/aid.1997.13.305

  126. Tramer MR, Carroll D, Campbell FA, Reynolds DJ, Moore RA, McQuay HJ (2001) Cannabinoids for control of chemotherapy induced nausea and vomiting: quantitative systematic review. Br Med J 323 (7303): 16–21

    Google Scholar 

  127. van den Elsen GA, Ahmed AI, Lammers M, Kramers C, Verkes RJ, van der Marck MA, Rikkert MG (2014) Efficacy and safety of medical cannabinoids in older subjects: a systematic review. Age Res Rev 14: 56–64. doi: 10.1016/j.arr.2014.01.007

  128. Whiting P, Savovic J, Higgins JP, Caldwell DM, Reeves BC, Shea B, Davies P, Kleijnen J, Churchill R (2016) ROBIS: A new tool to assess risk of bias in systematic reviews was developed. J Clin Epidemiol 69: 225-234. doi: 10.1016/j.jclinepi.2015.06.005

  129. Whiting PF, Wolff RF, Deshpande S, Di Nisio M, Duffy S, Hernandez AV, Keurentjes JC, Lang S, Misso K, Ryder S, Schmidlkofer S, Westwood M, Kleijnen J (2015) Cannabinoids for medical use: A systematic review and meta-analysis. J Am Med Assoc 313 (24): 2456–2473. doi: 10.1001/jama.2015.6358

  130. Yoodee J, Permsuwan U, Nimworapan M (2017) Efficacy and safety of olanzapine for the prevention of chemotherapy-induced nausea and vomiting: A systematic review and meta-analysis. Crit Rev Oncol Hematol 112: 113–125. doi: 10.1016/j.critrevonc.2017.02.017

  131. Allen KL, Waldvogel HJ, Glass M, Faull RL (2009) Cannabinoid (CB (1)), GABA (A) and GABA (B) receptor subunit changes in the globus pallidus in Huntington’s disease. J Chem Neuroanat 37 (4): 266–281

    Google Scholar 

  132. Ames FR, Cridland S (1985) Anticonvulsant effect of cannabidiol. S Afr Med J 14 (69)

    Google Scholar 

  133. Andrzejewski K, Barbano R, Mink J (2016) Cannabinoids in the treatment of movement disorders: A systematic review of case series and clinical trials. Basal Ganglia 6 (3): 173–181

    Google Scholar 

  134. Braida D, Pegorini S, Arcidiacono M, Consalez G, Croci L, Sala M (2003) Post-ischemic treatment with cannabidiol prevents electroencephalographic flattening, hyperlocomotion and neuronal injury in gerbils. Neurosci Lett (346): 61–64

    Google Scholar 

  135. Carroll C, Bain P, Teare L, Liu X, Joint C, Wroath C, Parkin S, Fox P, Wright D, Hobart J (2004) Cannabis for dyskinesia in Parkinson disease A randomized double-blind crossover study. Neurology 63 (7): 1245–1250

    Google Scholar 

  136. Chagas MHN, Zuardi AW, Tumas V, Pena-Pereira MA, Sobreira ET, Bergamaschi MM, dos Santos AC, Teixeira AL, Hallak JE, Crippa JAS (2014) Effects of cannabidiol in the treatment of patients with Parkinson’s disease: an exploratory double-blind trial. J Psychopharmacol 28 (11): 1088–1098

    Google Scholar 

  137. Collin C, Ehler E, Waberzinek G, Alsindi Z, Davies P, Powell K, Notcutt W, O’Leary C, Ratcliffe S, Nováková I, Zapletalova O, Piková J, Ambler Z (2010) A double-blind, randomized, placebo-controlled, parallel-group study of Sativex, in subjects with symptoms of spasticity due to multiple sclerosis. Neurol Res 32 (5): 451–459. doi: 10.1179/016164109X12590518685660

  138. Consroe P, Laguna J, Allender J, Snider S, Stern L, Sandyk R, Kennedy K, Schram K (1991) Controlled clinical trial of cannabidiol in Huntington’s disease. Pharmacol Biochem Behav 40 (3): 701–708

    Google Scholar 

  139. Cunha J, Carlini E, Pereira AE, Ramos OL, Pimentel C, Gagliardi R, Sanvito W, Lander N, Mechoulam R (1980) Chronic administration of cannabidiol to healthy volunteers and epileptic patients. Pharmacology 21 (3): 175–185

    Google Scholar 

  140. Curtis A, Mitchell I, Patel S, Ives N, Rickards H (2009) A pilot study using nabilone for symptomatic treatment in Huntington’s disease. Movement Disord 24 (15): 2254–2259

    Google Scholar 

  141. Davis JP, Ramsay HH (1994) Antiepileptic action of marijuana-active substances. Fed Proc (8): 284–285

    Google Scholar 

  142. Fox P, Bain P, Glickman S, Carroll C, Zajicek J (2004) The effect of cannabis on tremor in patients with multiple sclerosis. Neurology 62 (7): 1105–1109

    Google Scholar 

  143. Fox SH, Kellett M, Moore AP, Crossman AR, Brotchie JM (2002) Randomised, double-blind, placebo-controlled trial to assess the potential of cannabinoid receptor stimulation in the treatment of dystonia. Move Disord 17 (1): 145–149

    Google Scholar 

  144. Frankel JP, Hughes A, Lees AJ, Stern GM (1990) Marijuana for parkinsonian tremor. J Neurol Neurosurg Psychiat 53 (5): 436

    Google Scholar 

  145. Freeman R, Adekanmi O, Waterfield M, Waterfield A, Wright D, Zajicek J (2006) The effect of cannabis on urge incontinence in patients with multiple sclerosis: a multicentre, randomised placebo-controlled trial (CAMS-LUTS). Int Urogynecol J 17 (6): 636–641

    Google Scholar 

  146. Gloss D, Vickrey B (2014) Cannabinoids for epilepsy. Cochrane Database Syst Rev (3)

    Google Scholar 

  147. Izzo AA, Sharkey KA (2010) Cannabinoids and the gut: new developments and emerging concepts. Pharmacol Ther (126): 21–38

    Google Scholar 

  148. Jin KL, Mao X.O., Goldsmith PC, Greenberg DA (2000) CB1 cannabinoid receptor induction in experimental stroke. Ann Neurol (48): 257–261

    Google Scholar 

  149. Kavia R, De Ridder D, Constantinescu C, Stott C, Fowler C (2010) Randomized controlled trial of Sativex to treat detrusor overactivity in multiple sclerosis. Multiple Sclerosis J 16 (11): 1349–1359

    Google Scholar 

  150. Koppel BS, Brust JC, Fife T, Bronstein J, Youssof S, Gronseth G, Gloss D (2014) Systematic review: Efficacy and safety of medical marijuana in selected neurologic disorders – Report of the Guideline Development Subcommittee of the American Academy of Neurology. Neurology 82 (17): 1556–1563

    Google Scholar 

  151. Lewin S, Glenton C, Munthe-Kaas H, Carlsen B, Colvin CJ, Gülmezoglu M, Noyes J, Booth A, Garside R, Rashidian A (2015) Using Qualitative Evidence in Decision Making for Health and Social Interventions: An Approach to Assess Confidence in Findings from Qualitative Evidence Syntheses (GRADE-CERQual). PLOS Medicine 12 (10): e1001895. doi: 10.1371/journal.pmed.1001895

  152. López-Sendón JL, Caldentey JG, Trigo P, Ruiz C, Ribas GG, Aguilar M, Galvez MA, Pablo J, Galve-Roth I, Sagredo O, Fernandez-Ruiz JJ, Guzman M, García J (2012) A double blind, cross over, placebo-controlled, phase II trial of Sativex in Huntington’s disease. J Neurol Neurosurg Psychiat 83 (Supplement 1): A62

    Google Scholar 

  153. Lorenz R (2004) On the application of cannabis in paediatrics and epileptology. Neuroendocinol Lett (25): 40–44

    Google Scholar 

  154. Lotan I, Treves TA, Roditi Y, Djaldetti R (2014) Cannabis (medical marijuana) treatment for motor and non-motor symptoms of Parkinson disease: an openlabel observational study. Clin Neuropharmacol 37 (2): 41–44

    Google Scholar 

  155. Ludanyi A, Eross L, Czirjak S, Vajda J, Halasz P, Watanabe M, Palkovits M, Magloczky Z, Freund TF, Katona I (2008) Downregulation of the CB1 cannabinoid receptor and related molecular elements of the endocannabinoid system in epileptic human hippocampus. J Neurosci 28 (12): 2976–2990. doi: 10.1523/jneurosci.4465-07.2008

  156. Maione S, Costa B, Di Marzo V (2013) Endocannabinoids: A unique opportunity to develop multitarget analgesics. Pain 154: S87–S93.

    Google Scholar 

  157. Marsicano G, Moosmann B, Hermann H, Lutz B, Behl C (2002) Neuroprotective properties of cannabinoids against oxidative stress: role of the cannabinoid receptor CB1. J Neurochem 80 (3): 448–456. doi: 10.1046/j.0022-3042.2001.00716.x

  158. Mechoulam R, Carlini E (1978) Toward drugs derived from cannabis. Naturwissenschaften 65 (4): 174–179

    Google Scholar 

  159. Mesnage V, Houeto J, Bonnet A, Clavier I, Arnulf I, Cattelin F, Le Fur G, Damier P, Welter M, Agid Y (2004) Neurokinin B, neurotensin, and cannabinoid receptor antagonists and Parkinson disease. Clin Neuropharmacol 27 (3): 108–110

    Google Scholar 

  160. Naftali T, Mechulam R, Marii A, Gabay G, Stein A, Bronshtain M, Laish I, Benjaminov F, Konikoff FM (2017) Low-Dose Cannabidiol Is Safe but Not Effective in the Treatment for Crohn’s Disease, a Randomized Controlled Trial. Digest Dis Sci 62 (6): 1615–1620. doi: 10.1007/s10620-017-4540-z

  161. Naftali T, Schleider LB-L, Dotan I, Lansky EP, Benjaminov FS, Konikoff FM (2013) Cannabis induces a clinical response in patients with Crohn’s disease: a prospective placebo-controlled study. Clin Gastroenterol Hepatol 11 (10): 1276–1280. e1271

    Google Scholar 

  162. OCEBM (2011) The Oxford Levels of Evidence 2. Oxford Centre for Evidence-Based Medicine, 2 edn. http://www.cebm.net/index.aspx?o = 5653,

  163. Porter BE, Jacobson C (2013) Report of a parent survey of cannabidiol-enriched cannabis use in pediatric treatment-resistant epilepsy. Epilep Bahav (29): 574–577

    Google Scholar 

  164. Rodriguez-Cueto C, Hernandez-Galvez M, Hillard CJ, Maciel P, Garcia-Garcia L, Valdeolivas S, Pozo MA, Ramos JA, Gomez-Ruiz M, Fernandez-Ruiz J (2016) Dysregulation of the endocannabinoid signaling system in the cerebellum and brainstem in a transgenic mouse model of spinocerebellar ataxia type-3. Neuroscience 339: 191–209. doi: 10.1016/j.neuroscience.2016.09.046

  165. Romigi A, Bari M, Placidi F, Marciani MG, Malaponti M, Torelli F, Izzi F, Prosperetti C, Zannino S, Corte F, Chiaramonte C, Maccarrone M (2010) Cerebrospinal fluid levels of the endocannabinoid anandamide are reduced in patients with untreated newly diagnosed temporal lobe epilepsy. Epilepsia 51 (5): 768–772. doi: 10.1111/j.1528-1167.2009.02334.x

  166. Sieradzan K, Fox S, Hill M, Dick J, Crossman A, Brotchie J (2001) Cannabinoids reduce levodopa-induced dyskinesia in Parkinson’s disease: a pilot study. Neurology 57 (11): 2108–2111

    Google Scholar 

  167. SIGN (2015) Scottish Intercollegiate Guidelines Network, SIGN 50 Methodology Checklist. Edinburgh, United Kingdom

    Google Scholar 

  168. Sliwa JA, Bell HK, Mason KD, Gore RM, Nanninga J, Cohen B (1996) Upper urinary tract abnormalities in multiple sclerosis patients with urinary symptoms. Arch Phys Med Rehabil 77 (247): 68

    Google Scholar 

  169. Storr M, Devlin S, Kaplan GG, Panaccione R, Andrews CN (2014) Cannabis use provides symptom relief in patients with inflammatory bowel disease but is associated with worse disease prognosis in patients with Crohn’s disease. Inflammat Bowel Dis 20 (3): 472–480. doi: 10.1097/01.MIB.0000440982.79036.d6

  170. Thomasius R, Petersen KU (2007) Auwirkungen von Cannabiskonsum und -missbrauch. Eine Expertise zu gesundheitlichen und psychosozialen Folgen. 1 systematisches Review der international publizierten Studien von 1996–2006. Pabst Science Publishers, Lengerich

    Google Scholar 

  171. Tomida I, Azuara-Blanco A, House H, Flint M, Pertwee RG, Robson PJ (2006) Effect of sublingual application of cannabinoids on intraocular pressure: a pilot study. J Glaucoma 15 (5): 349–353

    Google Scholar 

  172. Trembly B, Sherman M Double-blind clinical study of cannabidiol as a secondary anticonvulsant. In: Marijuana „90 International Conference on Cannabis and Cannabinoids, Kolympari, Crete., 1990. vol section 2. International Association for Cannabinoid Medicines, p 5

    Google Scholar 

  173. Van Laere K, Casteels C, Dhollander I, Goffin K, Grachev I, Bormans G, Vandenberghe W (2010) Widespread decrease of type 1 cannabinoid receptor availability in Huntington disease in vivo. J Nucl Med 51 (9): 1413–1417. doi: 10.2967/jnumed.110.077156

  174. Vaney C, Heinzel-Gutenbrunner M, Jobin P, Tschopp F, Gattlen B, Hagen U, Schnelle M, Reif M (2004) Efficacy, safety and tolerability of an orally administered cannabis extract in the treatment of spasticity in patients with multiple sclerosis: a randomized, double-blind, placebo-controlled, crossover study. Multiple Sclerosis 10 (4): 417–424

    Google Scholar 

  175. Volz M, Siegmund B, Häuser PDW (2016) Wirksamkeit, Verträglichkeit und Sicherheit von Cannabinoiden in der Gastroenterologie. Schmerz 30 (1): 37–46

    Google Scholar 

  176. Wade DT, Makela P, Robson P, House H, Bateman C (2004) Do cannabis-based medicinal extracts have general or specific effects on symptoms in multiple sclerosis? A double-blind, randomized, placebo-controlled study on 160 patients. Multiple Sclerosis 10 (4): 434–441

    Google Scholar 

  177. Whiting P, Savovic J, Higgins JP, Caldwell DM, Reeves BC, Shea B, Davies P, Kleijnen J, Churchill R (2016) ROBIS: A new tool to assess risk of bias in systematic reviews was developed. J Clin Epidemiol 69: 225–234. doi: 10.1016/j.jclinepi.2015.06.005

  178. Whiting PF, Wolff RF, Deshpande S, Di Nisio M, Duffy S, Hernandez AV, Keurentjes JC, Lang S, Misso K, Ryder S (2015) Cannabinoids for medical use: a systematic review and meta-analysis. Jama 313 (24): 2456–2473

    Google Scholar 

  179. Wong BS, Camilleri M, Eckert D, Carlson P, Ryks M, Burton D, Zinsmeister AR (2012) Randomized pharmacodynamic and pharmacogenetic trial of dronabinol effects on colon transit in irritable bowel syndrome-diarrhea. Neurogastroenterol Motil 24 (4): 358-e169

    Google Scholar 

  180. Yap M, Easterbrook L, Connors J, Koopmans L (2015) Use of cannabis in severe childhood epilepsy and child protection considerations. J Paediat Child Health 51 (5): 491–496. doi: http://dx.doi.org/10.1111/jpc.12876

  181. Zadikoff C, Wadia P, Miyasaki J, Chen R, Lang A, So J, Fox S (2011) Cannabinoid, CB1 agonists in cervical dystonia: failure in a phase IIa randomized controlled trial. Basal Ganglia 1 (2): 91–95

    Google Scholar 

  182. Zajicek JP, Fox P, Sanders H, al; e, Group. UMR (2003) Cannabinoids for treatment of spasticity and other symptoms related to multiple sclerosis (CAMS study): multicentre randomised placebo-controlled trial. Lancet (362): 1517–1526.

    Google Scholar 

  183. Zuardi AW, Crippa JA, Hallak JE, Pinto JP, Chagas MH, Rodrigues GG, Dursun SM, Tumas V (2009) Cannabidiol for the treatment of psychosis in Parkinson’s disease. J Psychopharmacol (Oxf) 23 (8): 979–983

    Google Scholar 

  184. Ahmed AI, van den Elsen GA, Colbers A, Kramers C, Burger DM, van der Marck MA, Olde Rikkert MG (2015) Safety, pharmacodynamics, and pharmacokinetics of multiple oral doses of delta-9-tetrahydrocannabinol in older persons with dementia. Psychopharmacology 232 (14): 2587–2595. doi: 10.1007/s00213-015-3889-y

  185. Andries A, Frystyk J, Flyvbjerg A, Stoving RK (2015a) Changes in IGF-I, urinary free cortisol and adipokines during dronabinol therapy in anorexia nervosa: Results from a randomised, controlled trial. Growth Hormone IGF Res 25 (5): 247–252. doi: 10.1016/j.ghir.2015.07.006

  186. Andries A, Frystyk J, Flyvbjerg A, Støving RK (2014) Dronabinol in severe, enduring anorexia nervosa: A randomized controlled trial. Int J Eat Disord 47 (1): 18–23. doi: 10.1002/eat.22173

  187. Andries A, Gram B, Stoving RK (2015b) Effect of dronabinol therapy on physical activity in anorexia nervosa: a randomised, controlled trial. Eat Weight Disord 20 (1): 13–21. doi: http://dx.doi.org/10.1007/s40519-014-0132-5

  188. Andrzejewski K, Barbano R, Mink J (2016) Cannabinoids in the treatment of movement disorders: A systematic review of case series and clinical trials. Basal Ganglia 6 (3): 173–181. doi: http://dx.doi.org/10.1016/j.baga.2016.06.001

  189. Bergamaschi MM, Queiroz RHC, Chagas MHN, de Oliveira DCG, De Martinis BS, Kapczinski F, …, Crippa JAS (2011) Cannabidiol Reduces the Anxiety Induced by Simulated Public Speaking in Treatment-Naïve Social Phobia Patients. Neuropsychopharmacology 36 (6): 1219–1226.

    Google Scholar 

  190. Bisaga A, Sullivan MA, Glass A, Mishlen K, Pavlicova M, Haney M, Raby WN, Levin FR, Carpenter KM, Mariani JJ, Nunes EV (2015) The effects of dronabinol during detoxification and the initiation of treatment with extended release naltrexone. Drug Alcohol Depend 154: 38–45. doi: 10.1016/j.drugalcdep.2015.05.013

  191. Bisogno T, MacCarrone M, De Petrocellis L, Jarrahian A, FinazziAgro A, Hillard C, Di Marzo V (2001) The uptake by cells of 2arachidonoylglycerol, an endogenous agonist of cannabinoid receptors. Eur J Biochem 268: 1982–1989

    Google Scholar 

  192. Boggs DL, Kelly DL, McMahon RP, Gold JM, Gorelick DA, Linthicum J, et al. (2012) Rimonabant for neurocognition in schizophrenia: A 16-week double blind randomized placebo controlled trial. Schizophr Res 134: 207–210

    Google Scholar 

  193. Frank B, Serpell MG, Hughes J, Matthews JNS, Kapur D (2008) Comparison of analgesic effects and patient tolerability of nabilone and dihydrocodeine for chronic neuropathic pain: randomised, crossover, double blind study. Br Med J 336 (7637): 199–201

    Google Scholar 

  194. Hay P, Touyz S (2015) Treatment of patients with severe and enduring eating disorders. Curr Opin Psychiat 28 (6): 473–477. doi: http://dx.doi.org/10.1097/YCO.0000000000000191

  195. Hill MN, Tasker JG (2012) Endocannabinoid signaling, glucocorticoid-mediated negative feedback, and regulation of the hypothalamic-pituitary-adrenal axis. Neuroscience 204: 5–16. doi: 10.1016/j.neuroscience.2011.12.030

  196. Iseger TA, Bossong MG (2015) A systematic review of the antipsychotic properties of cannabidiol in humans. Schizophr Res 162 (1–3): 153–161. doi: http://dx.doi.org/10.1016/j.schres.2015.01.033

  197. Jetly R, Heber A, Fraser G, Boisvert D (2015) The efficacy of nabilone, a synthetic cannabinoid, in the treatment of PTSD-associated nightmares: A preliminary randomized, double-blind, placebo-controlled cross-over design study. Psychoneuroendocrinology 51: 585–588. doi: http://dx.doi.org/10.1016/j.psyneuen.2014.11.002

  198. Jicha CJ, Lofwall MR, Nuzzo PA, Babalonis S, Elayi SC, Walsh SL (2015) Safety of oral dronabinol during opioid withdrawal in humans. Drug Alcohol Depend 157: 179–183. doi: 10.1016/j.drugalcdep.2015.09.031

  199. Koppel BS, Brust JCM, Fife T, Bronstein J, Youssof S, Gronseth G, Gloss D (2014) Systematic review: Efficacy and safety of medical marijuana in selected neurologic disorders: Report of the Guideline Development Subcommittee of the American Academy of Neurology. Neurology 82 (17): 1556–1563. doi: 10.1212/WNL.0000000000000363

  200. Krishnan S, Cairns R, R. H (2009) Cannabinoids for the treatment of dementia. Cochrane Database Syst Rev (Issue 2: CD007204. doi: 10.1002/14651858.CD007204.pub2

  201. Levin FR, Mariani JJ, Pavlicova M, Brooks D, Glass A, Mahony A, Nunes EV, Bisaga A, Dakwar E, Carpenter KM, Sullivan MA, Choi JC (2016) Dronabinol and lofexidine for cannabis use disorder: A randomized, double-blind, placebo-controlled trial. Drug Alcohol Depend 159: 53–60. doi: http://dx.doi.org/10.1016/j.drugalcdep.2015.11.025

  202. Leweke FM, Koethe D (2008) Cannabis and psychiatric disorders: it is not only addiction. Addict Biol 13 (2): 264–275. doi: 10.1111/j.1369-1600.2008.00106.x

  203. Leweke FM, Mueller JK, Lange B, Rohleder C (2016) Therapeutic potential of cannabinoids in psychosis. Biol Psychiat 79 (7): 604–612. doi: http://dx.doi.org/10.1016/j.biopsych.2015.11.018

  204. Leweke FM, Piomelli D, Pahlisch F, Muhl D, Gerth CW, Hoyer C, al. e (2012) Cannabidiol enhances anandamide signalling and alleviates psychotic symptoms of schizophrenia. Translat Psychiat 94: 1–7

    Google Scholar 

  205. Lewin S, Glenton C, Munthe-Kaas H, Carlsen B, Colvin CJ, Gülmezoglu M, Noyes J, Booth A, Garside R, Rashidian A (2015) Using Qualitative Evidence in Decision Making for Health and Social Interventions: An Approach to Assess Confidence in Findings from Qualitative Evidence Syntheses (GRADE-CERQual). PLOS Medicine 12 (10): e1001895. doi: 10.1371/journal.pmed.1001895

  206. Marco EM, Garcia-Gutierrez MS, Bermudez-Silva FJ, Moreira FA, Guimaraes F, Manzanares J, Viveros MP (2011) Endocannabinoid system and psychiatry: in search of a neurobiological basis for detrimental and potential therapeutic effects. Front Behav Neurosci 5: 63. doi: 10.3389/fnbeh.2011.00063

  207. McLoughlin BC (2014) Cannabis and schizophrenia. Cochrane Database Syst Rev 10: CD004837

    Google Scholar 

  208. Meltzer HY, Arvanitis L, Bauer D, Rein W (2004) Placebo-controlled evaluation of four novel compounds for the treatment of schizophrenia and schizoaffective disorder. Am J Psychiat 161: 975–984

    Google Scholar 

  209. Mücke M, Carter C, Cuhls H, Prüss M, Radbruch L, Häuser W (2016) Cannabinoids in palliative care: Systematic review and meta-analysis of efficacy, tolerability and safety. [German] (Cannabinoide in der palliativen Versorgung: Systematische Ubersicht und Metaanalyse der Wirksamkeit, Vertraglichkeit und Sicherheit.). Schmerz (Berlin, Germany) 30 (1): 25–36. doi: http://dx.doi.org/10.1007/s00482-015-0085-2

  210. Müller-Vahl KR, Koblenz A, Jöbges M, Kolbe H, Emrich HM, Schneider U (2001) Influence of treatment of Tourette syndrome with delta9tetrahydrocannabinol (delta9-THC) on neuropsychological performanc. Pharmacopsychiatry 34 (1): 19–24

    Google Scholar 

  211. Müller-Vahl KR, Prevedel H, Theloe K, Kolbe H, Emrich HM, U. S (2003a) Treatment of Tourette syndrome with delta-9-tetrahydrocannabinol (delta 9-THC): no influence on neuropsychological performance. Neuropsychopharmacology 28 (2): 384–388

    Google Scholar 

  212. Müller-Vahl KR, Schneider U, Koblenz A, al. e (2002) Treatment of Tourette’s syndrome with Delta 9-tetrahydrocannabinol (THC): a randomized crossover trial. Pharmacopsychiatry 35 (2): 57–61

    Google Scholar 

  213. Muller-Vahl KR, Schneider U, Koblenz A, Jobges M, Kolbe H, Daldrup T, Emrich HM (2002) Treatment of Tourette’s syndrome with Delta 9-tetrahydrocannabinol (THC): a randomized crossover trial. Pharmacopsychiatry 35 (2): 57–61. doi: 10.1055/s-2002-25028

  214. Müller-Vahl KR, Schneider U, Prevedel H, al. e (2003b) Delta 9-tetrahydrocannabinol (THC) is effective in the treatment of tics in Tourette syndrome: a 6-week randomized trial. J Clin Psychiat 64 (4): 459–465

    Google Scholar 

  215. Muller-Vahl KR, Schneider U, Prevedel H, Theloe K, Kolbe H, Daldrup T, Emrich HM (2003) Delta 9-tetrahydrocannabinol (THC) is effective in the treatment of tics in Tourette syndrome: a 6-week randomized trial. J Clin Psychiat 64 (4): 459–465

    Google Scholar 

  216. Narang S, Gibson D, Wasan AD, al. e (2008) Efficacy of dronabinol as an adjuvant treatment for chronic pain patients on opioid therapy. J Pain 9 (3): 254–264

    Google Scholar 

  217. OCEBM (2011) The Oxford Levels of Evidence 2. Oxford Centre for Evidence-Based Medicine, 2 edn. http://www.cebm.net/index.aspx?o = 5653,

  218. Parsons LH, Hurd YL (2015) Endocannabinoid signalling in reward and addiction. Nature Rev Neurosci 16 (10): 579–594. doi: 10.1038/nrn4004

  219. Portenoy RK, Ganae-Motan ED, Allende S, Yanagihara R, Shaiova L, Weinstein S, McQuade R, Wright S, Fallon MT (2012) Nabiximols for opioid-treated cancer patients with poorly-controlled chronic pain: a randomized, placebo-controlled, graded-dose trial. J Pain 13 (5): 438–449

    Google Scholar 

  220. Rog DJ, Nurmikko TJ, Friede T, Young CA (2005) Randomized, controlled trial of cannabis-based medicine in central pain in multiple sclerosis. Neurology 65: 812–819

    Google Scholar 

  221. Rohleder C, Müller JK, Lange B, Leweke FM (2016) Cannabidiol as a Potential New Type of an Antipsychotic. A Critical Review of the Evidence. Frontiers Pharmacol 7 (422). doi: http://doi.org/10.3389/fphar.2016.00422

  222. Rohleder C, Pahlisch F, Schaefer C, al. e (2012) The endocannabinoid system as a pharmacological target for antipsychotic treatment and more? Paper presented at 8th International Conference on Early Psychosis: From Neurobiology to Public Policy; 11–13 Oct 2012; San Francisco: CA. Early Interventions in Psychiatry 6 (Suppl 1): 7

    Google Scholar 

  223. Rubino T, Zamberletti E, Parolaro D (2015) Endocannabinoids and mental disorders. Handb Exp Pharmacol 231: 261–283. doi: 10.1007/978-3-319-20825-1_9

  224. Sanofi (2009) Efficacy and Safety of AVE1625 as a Co-treatment With Antipsychotic Therapy in Schizophrenia (CONNECT). Available at: http://ClinicalTrialsgov/show/NCT00439634 NLM Identifier: NCT00439634 Accessed March 31, 2015

  225. Schaefer C, Enning F, Mueller JK, Bumb JM, Rohleder C, Odorfer TM, Klosterkotter J, Hellmich M, Koethe D, Schmahl C, Bohus M, Leweke FM (2014) Fatty acid ethanolamide levels are altered in borderline personality and complex posttraumatic stress disorders. Eur Arch Psychiat Clin Neurosci 264 (5): 459–463. doi: 10.1007/s00406-013-0470-8

  226. Schubart CD, I.E.C. Sommera IEC, Fusar-Polib P, de Wittea L, Kahnc RS, Boksa MPM (2014) Cannabidiol as a potential treatment for psychosis. Eur Neuropsychopharmacol 24: 51–64

    Google Scholar 

  227. Schwarcz G, Karajgi B, McCarthy R (2009) Synthetic delta-9-tetrahydrocannabinol (dronabinol) can improve the symptoms of schizophrenia. J Clin Psychopharmacol 29: 255–258

    Google Scholar 

  228. SIGN (2015) Scottish Intercollegiate Guidelines Network. SIGN 50 Methodology Checklist. Edinburgh, United Kingdom

    Google Scholar 

  229. Steinberg K, Roffman R, Carroll K, McRee B, Babor T, Miller M, Kadden R, Duresky D, Stephens R (2005) Brief Counseling for Marijuana Dependence. Center for Substance Abuse Treatment, Substance Abuse and Mental Health Services Administration; Rockville, MD

    Google Scholar 

  230. Thomasius R, Petersen KU (2007) Auwirkungen von Cannabiskonsum und -missbrauch. Eine Expertise zu gesundheitlichen und psychosozialen Folgen. Ein systematisches Review der international publizierten Studien von 1996–2006. Pabst Science Publishers, Lengerich

    Google Scholar 

  231. Trigo JM, Lagzdins D, Rehm J, Selby P, Gamaleddin I, Fischer B, Barnes AJ, Huestis MA, Le Foll B (2016) Effects of fixed or self-titrated dosages of Sativex on cannabis withdrawal and cravings. Drug Alcohol Depend 161: 298–306. doi: 10.1016/j.drugalcdep.2016.02.020

  232. van den Elsen GA, Ahmed AI, Verkes RJ, Feuth T, van der Marck MA, Olde Rikkert MG (2015a) Tetrahydrocannabinol in Behavioral Disturbances in Dementia: A Crossover Randomized Controlled Trial. Am J Geriat Psychiat 23 (12): 1214–1224. doi: 10.1016/j.jagp.2015.07.011

  233. van den Elsen GAH, Ahmed AIA, Verkes R-J, Kramers C, Feuth T, Rosenberg PB, van der Marck MA, Olde Rikkert MGM (2015b) Tetrahydrocannabinol for neuropsychiatric symptoms in dementia: A randomized controlled trial. Neurology 84 (23): 2338–2346. doi: 10.1212/WNL.0000000000001675

  234. Volicer L, Stelly M, Morris J, McLaughlin J, Volicer BJ (1997) Effects of Dronabinol on anorexia and disturbed behavior in patients with Alzheimer’s disease. Int J Geriat Psychiat 12: 913–919

    Google Scholar 

  235. Wade DT, Makela P, Robson P, House H, Bateman C (2004) Do cannabis-based medicinal extracts have general or specific effects on symptoms in multiple sclerosis? A double-blind, randomized, placebo-controlled study on 160 patients. Multiple Sclerosis 10 (4): 434–441

    Google Scholar 

  236. Whiting PF, Wolff RF, Deshpande S, Di Nisio M, Duffy S, Hernandez AV, Keurentjes JC, Lang S, Misso K, Ryder S, Schmidlkofer S, Westwood M, Kleijnen J (2015) Cannabinoids for medical use: A systematic review and meta-analysis. J Am Med Assoc 313 (24): 2456–2473. doi: 10.1001/jama.2015.6358

  237. Whiting P, Savovic J, Higgins JP, Caldwell DM, Reeves BC, Shea B, Davies P, Kleijnen J, Churchill R (2016) ROBIS: A new tool to assess risk of bias in systematic reviews was developed. J Clin Epidemiol 69: 225–234. doi: 10.1016/j.jclinepi.2015.06.005

  238. Yarnell S (2015) The use of medicinal marijuana for posttraumatic stress disorder: A review of the current literature. Primary Care Companion to the J Clin Psychiat 17 (3). doi: http://dx.doi.org/10.4088/PCC.15r01786

  239. Zuardi AW, Hallak JE, Dursun SM, Morais SL, Faria Sanches R, Musty RE, Crippa JA (2006) Cannabidiol monotherapy for treatment-resistant schizophrenia. J Psychopharmacol 20: 683–686

    Google Scholar 

  240. Zuardi AW, Morais SL, Guimaraes FS, Mechoulam R (1995) Antipsychotic effect of cannabidiol. J Clin Psychiat 56: 485–486

    Google Scholar 

  241. APA (American Psychiatric Association) (2000) Diagnostic and statistical manual of mental disorders (4th edn, text rev.). American Psychiatric Association. doi: 10.1176/appi.books.9780890423349.

  242. BAH (2013) https://www.bah-bonn.de/unsere-themen/selbstmedikation. Bundesfachverband der Arzneimittelhersteller (Stand: 24.Sept 24 2017)

  243. Buadze A, Kaiser S, Stohler R, Roessler W, Seifritz E, Liebrenz M (2012) Patient’s perceptions of the cannabis-psychosis link--a systematic review. Curr Pharmaceut Design 18 (32): 5105–5112

    Google Scholar 

  244. Buadze A, Stohler R, Schulze B, Schaub M, Liebrenz M (2010) Do patients think cannabis causes schizophrenia? - A qualitative study on the causal beliefs of cannabis using patients with schizophrenia. Harm Reduct J 7: 22. doi: 10.1186/1477-7517-7-22

  245. Buckner JD, Ecker AH, Vinci C (2013) Cannabis use vulnerability among socially anxious users: cannabis craving during a social interaction. Psychol Addict Behav 27 (1): 236–242. doi: 10.1037/a0029763

  246. Buckner JD, Jeffries ER, Terlecki MA, Ecker AH (2016) Distress tolerance among students referred for treatment following violation of campus cannabis use policy: Relations to use, problems, and motivation. Behav Modif 40 (5): 663–677. doi: 10.1177/0145445515610315

  247. Dilling H, Mombour W, Schmidt MH (2010) Internationale Klassifikation psychischer Störungen. ICD-10. Huber, Bern

    Google Scholar 

  248. Feingold (2015) The association between cannabis use and anxiety disorders: Results from a population-based representative sample. Eur Neuropsychopharmacol 26, 3: 493–505

    Google Scholar 

  249. Feingold D, Weiser M, Rehm J, Lev-Ran S (2015) The association between cannabis use and mood disorders: A longitudinal study. J Affect Disord 172: 211–218

    Google Scholar 

  250. Feingold D, Weiser M, Rehm J, Lev-Ran S (2016) The association between cannabis use and anxiety disorders: Results from a population-based representative sample. Eur Neuropsychopharmacol 26 (3): 493–505. doi: 10.1016/j.euroneuro.2015.12.037

  251. Fox CL, Towe SL, Stephens RS, Walker DD, Roffman RA (2011) Motives for cannabis use in high-risk adolescent users. Psychol Addict Behav 25 (3): 492–500. doi: 10.1037/a0024331

  252. Gill KE, Poe L, Azimov N, Ben-David S, Vadhan NP, Girgis R, Moore H, Cressman V, Corcoran CM (2015) Reasons for cannabis use among youths at ultra high risk for psychosis. Early Entervent Psychiat 9 (3): 207–210. doi: 10.1111/eip.12112

  253. Gomez Pérez L, Santacana AM, Berge Baquero D, Perez-Sola V (2014) Reasons and subjective effects of cannabis use among people with psychotic disorders: a systematic review. Actas Esp Psiquiatr 42 (2): 83–90

    Google Scholar 

  254. Goswami S, Mattoo SK, Basu D, Singh G (2004) Substance-abusing schizophrenics: do they self-medicate? Am J Addiction (13): 139–150

    Google Scholar 

  255. Grant BF, Kaplan KD (2005) Source and Accuracy Statement for the 2004–2005 Wave 2 National Epidemiologic Survey on Alcohol and Related Conditions. Bethesda, MD

    Google Scholar 

  256. Grant BF, Kaplan KD, Shepard J, Moore T (2003) Source and Accuracy Statement for Wave 1 of the 2001–2002 National Epidemiologic Survey on Alcohol and Related Conditions. Bethesda, MD

    Google Scholar 

  257. Green B, Kavanagh DJ, Young RM (2004) Reasons for cannabis use in men with and without psychosis. Drug Alcohol Rev 23 (4): 445–453. doi: 10.1080/09595230412331324563

  258. Grinspoon L, Bakalar JB (2000) Marihuana. Die verbotene Medizin. Zweitausenens, Frankfurt

    Google Scholar 

  259. Kolliakou A, Castle D, Sallis H, Joseph C, O’Connor J, Wiffen B, Gayer-Anderson C, McQueen G, Taylor H, Bonaccorso S, Gaughran F, Smith S, Greenwood K, Murray RM, Di Forti M, Atakan Z, Ismail K (2015) Reasons for cannabis use in first-episode psychosis: does strength of endorsement change over 12 months? Eur Psychiat 30 (1): 152–159. doi: 10.1016/j.eurpsy.2014.10.007

  260. Lewin S, Glenton C, Munthe-Kaas H, Carlsen B, Colvin CJ, Gülmezoglu M, Noyes J, Booth A, Garside R, Rashidian A (2015) Using Qualitative Evidence in Decision Making for Health and Social Interventions: An Approach to Assess Confidence in Findings from Qualitative Evidence Syntheses (GRADE-CERQual). PLOS Medicine 12 (10): e1001895. doi: 10.1371/journal.pmed.1001895

  261. Mané A, Fernández-Expósito M, Bergé D, Gómez-Pérez L, Sabaté A, Toll A, Diaz L, Diez-Aja C, Perez V (2015) Relationship between cannabis and psychosis: Reasons for use and associated clinical variables. Psychiat Res 229 (1–2): 70–74. doi: 10.1016/j.psychres.2015.07.070

  262. OCEBM (2011) Levels of Evidence Working Group. The Oxford Levels of Evidence 2. http://www.cebm.net/index.aspx?o = 5653, 2 edn. Oxford Centre for Evidence-Based Medicine, Oxford

  263. Roulette CJ, Kazanji M, Breurec S, Hagen EH (2016) High prevalence of cannabis use among Aka foragers of the Congo Basin and its possible relationship to helminthiasis. Am J Human Biol 28 (1): 5–15. doi: 10.1002/ajhb.22740

  264. Schaub M, Fanghaenel K, Stohler R (2008) Reasons for cannabis use: patients with schizophrenia versus matched healthy controls. Aust N Z J Psychiat 42 (12): 1060–1065. doi: 10.1080/00048670802512016

  265. Schofield D, Tennant C, Nash L, Degenhardt L, Cornish A, Hobbs C, Brennan G (2006) Reasons for cannabis use in psychosis. Aust NZ J Psychiat 40 (6–7): 570–574. doi: 10.1080/j.1440-1614.2006.01840.x

  266. SIGN (2015) Scottish Intercollegiate Guidelines Network. SIGN 50 Methodology Checklist. Edinburgh, United Kingdom

    Google Scholar 

  267. Simons J, Correia C, Carey K, Borsari B (1998) Validating a five-factor marijuana motives measure: Relations with use, problems, and alcohol motives. J Counsel Psychol 45: 265–273

    Google Scholar 

  268. Thomasius R, Petersen KU (2007) Auwirkungen von Cannabiskonsum und -missbrauch. Eine Expertise zu gesundheitlichen und psychosozialen Folgen. 1 systematisches Review der international publizierten Studien von 1996–2006. Pabst Science Publishers, Lengerich

    Google Scholar 

  269. Tyler E, Jones A, Black N, Carter L-A, Barrowclough C (2015) The Relationship between Bipolar Disorder and Cannabis Use in Daily Life: An Experience Sampling Study. PloS one 10 (3). doi: e0118916. pmid: 25738578

    Google Scholar 

  270. Wadley G (2016) How psychoactive drugs shape human culture: A multi-disciplinary perspective. Brain Res Bull 126 (Pt 1): 138–151. doi: 10.1016/j.brainresbull.2016.04.008

  271. Whiting P, Savović J, Higgins JPT, Caldwell DM, Reeves BC, Shea B, Davies P, Kleijnen J, Churchill R, group aR (2016) ROBIS: A new tool to assess risk of bias in systematic reviews was developed. J Clin Epidemiol 69: 225–234

    Google Scholar 

  272. Whiting PF, Wolff RF, Deshpande S, Di Nisio M, Duffy S, Hernandez AV, Keurentjes JC, Lang S, Misso K, Ryder S, Schmidlkofer S, Westwood M, Kleijnen J (2015) Cannabinoids for medical use: A systematic review and meta-analysis. J Am Med Assoc 313 (24): 2456–2473. doi: 10.1001/jama.2015.6358

Download references

Author information

Authors and Affiliations

Authors

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2019 Springer-Verlag GmbH Deutschland, ein Teil von Springer Nature

About this chapter

Cite this chapter

Hoch, E. et al. (2019). Wirksamkeit, Verträglichkeit und Sicherheit von medizinischem Cannabis. In: Hoch, E., Friemel, C.M., Schneider, M. (eds) Cannabis: Potenzial und Risiko. Springer, Berlin, Heidelberg. https://doi.org/10.1007/978-3-662-57291-7_4

Download citation

  • DOI: https://doi.org/10.1007/978-3-662-57291-7_4

  • Publisher Name: Springer, Berlin, Heidelberg

  • Print ISBN: 978-3-662-57290-0

  • Online ISBN: 978-3-662-57291-7

  • eBook Packages: Medicine (German Language)

Publish with us

Policies and ethics