Effect of Inhaled Cannabis for Pain in Adults With Sickle Cell Disease: A Randomized Clinical Trial

Donald I Abrams, Paul Couey, Niharika Dixit, Varun Sagi, Ward Hagar, Elliott Vichinsky, Mary Ellen Kelly, John E Connett, Kalpna Gupta, Donald I Abrams, Paul Couey, Niharika Dixit, Varun Sagi, Ward Hagar, Elliott Vichinsky, Mary Ellen Kelly, John E Connett, Kalpna Gupta

Abstract

Importance: Sickle cell disease (SCD) is characterized by chronic pain and episodic acute pain caused by vasoocclusive crises, often requiring high doses of opioids for prolonged periods. In humanized mouse models of SCD, a synthetic cannabinoid has been found to attenuate both chronic and acute hyperalgesia. The effect of cannabis on chronic pain in adults with SCD is unknown.

Objective: To determine whether inhaled cannabis is more effective than inhaled placebo in relieving chronic pain in adults with SCD.

Design, setting, and participants: This pilot randomized clinical trial included participants with SCD with chronic pain admitted to a single inpatient clinical research center for 2 separate 5-day stays from August 2014 to April 2017. Participants inhaled either vaporized cannabis (4.4% Δ-9-tetrahydrocannabinol to 4.9% cannabidiol) 3 times daily or vaporized placebo cannabis. Pain and pain interference ratings using the Brief Pain Inventory were assessed throughout each 5-day period. Participants with SCD and chronic pain on stable analgesics were eligible to enroll. A total of 90 participants were assessed for eligibility; 56 participants were deemed ineligible, and 34 participants were enrolled. Of these, 7 participants dropped out before randomization. Of 27 randomized participants, 23 completed both treatment arms of the crossover study and were included in the final per protocol analysis. Data analysis was completed in June 2019, with the sensitivity analysis conducted in April 2020.

Interventions: Inhalation of vaporized cannabis plant (4.4% Δ-9-tetrahydrocannbinol to 4.9% cannabidiol) or placebo cannabis plant using a vaporizer 3 times daily for 5 days.

Main outcomes and measures: Daily pain assessed with visual analog scale and Brief Pain Inventory.

Results: A total of 23 participants (mean [SD] age, 37.6 [11.4] years; 13 [56%] women) completed the trial. The mean (SD) difference in pain rating assessment between the cannabis and placebo groups was -5.3 (8.1) for day 1, -10.9 (7.0) for day 2, -16.5 (9.2) for day 3, -8.9 (6.7) for day 4, and -8.2 (8.1) for day 5; however, none of these differences were statistically significant. There was no statistically significant mean (SD) difference in pain interference ratings between cannabis and placebo between days 1 and 5 for interference in general activities (day 1: 0.27 [0.35]; day 5: -1.0 [0.5]), walking (day 1: 0.14 [0.73]; day 5: -0.87 [0.63]), sleep (day 1: 0.59 [0.74]; day 5: -1.3 [0.8]), or enjoyment (day 1: 0.23 [0.69]; day 5: -0.91 [0.48]), but there was a statistically significant mean (SD) difference in decrease in interference with mood (day 1: 0.96 [0.59]; day 5: -1.4 [0.6]; P = .02). No differences in treatment-related adverse effects were observed. Use of concomitant opioids was similar during both treatment periods.

Conclusions and relevance: This randomized clinical trial found that, compared with vaporized placebo, vaporized cannabis did not statistically significantly reduce pain and associated symptoms, except interference in mood, in patients with SCD with chronic pain.

Trial registration: ClinicalTrials.gov Identifier: NCT01771731.

Conflict of interest statement

Conflict of Interest Disclosures: Dr Abrams reported receiving personal fees from AXIM Biotechnologies, INSYS, Intec, Maui Wellness Group, MedLab Clinical Ltd, Scriptyx, Spectrum Cannabis, Tikun Olam, and VIVO Cannabis; and stock ownership in Cannformatics and Lumen outside the submitted work. Dr Connett reported receiving grants from the National Institutes of Health (NIH) and US Department of Defense outside the submitted work. Dr Gupta reported receiving grants from 1910 Genetics, Grifols, the Southern California Institute for Research and Education Foundation, and the University of Minnesota Foundation; personal fees from Tautona Group, Novartis, and CSL Behring; and serving as a board member of the Sickle Cell Disease Association of America outside the submitted work. No other disclosures were reported.

Figures

Figure 1.. Consort Diagram
Figure 1.. Consort Diagram
Figure 2.. Daily Pain Rating During Cannabis…
Figure 2.. Daily Pain Rating During Cannabis and Placebo Treatment
Participants completed a self-reported assessment of pain on the Drug Effects Questionnaire during each day of cannabis and placebo treatment. Pain was rated on a visual analog scale from 0 to 100. Dots indicate mean and whiskers, 95% CI.
Figure 3.. Difference in Pain Interference Ratings…
Figure 3.. Difference in Pain Interference Ratings Between Cannabis and Placebo Treatment
The brief pain inventory, a self-reported assessment of various chronic pain interference measures, was completed by each patient on days 1 and 5 of the cannabis and placebo treatment periods. Each of the interference criteria was rated on a scale from 1 to 10. Dots indicate mean and whiskers, 95% CI.

References

    1. National Conference of State Legislatures State medical marijuana laws. Accessed May 31, 2019.
    1. Kosiba JD, Maisto SA, Ditre JW. Patient-reported use of medical cannabis for pain, anxiety, and depression symptoms: systematic review and meta-analysis. Soc Sci Med. 2019;233:181-192. doi:10.1016/j.socscimed.2019.06.005
    1. National Academies of Sciences, Engineering and Medicine The Health Effects of Cannabis and Cannabinoids: The Current State of Evidence and Recommendations for Research. National Academies Press; 2017.
    1. Niihara Y, Miller ST, Kanter J, et al. ; Investigators of the Phase 3 Trial of l-Glutamine in Sickle Cell Disease . A phase 3 trial of l-glutamine in sickle cell disease. N Engl J Med. 2018;379(3):226-235. doi:10.1056/NEJMoa1715971
    1. Ataga KI, Kutlar A, Kanter J, et al. . Crizanlizumab for the prevention of pain crises in sickle cell disease. N Engl J Med. 2017;376(5):429-439. doi:10.1056/NEJMoa1611770
    1. Smith WR, Penberthy LT, Bovbjerg VE, et al. . Daily assessment of pain in adults with sickle cell disease. Ann Intern Med. 2008;148(2):94-101. doi:10.7326/0003-4819-148-2-200801150-00004
    1. Ruta NS, Ballas SK. The opioid drug epidemic and sickle cell disease: guilt by association. Pain Med. 2016;17(10):1793-1798. doi:10.1093/pm/pnw074
    1. Brandow AM, Farley RA, Panepinto JA. Early insights into the neurobiology of pain in sickle cell disease: a systematic review of the literature. Pediatr Blood Cancer. 2015;62(9):1501-1511. doi:10.1002/pbc.25574
    1. Tran H, Gupta M, Gupta K. Targeting novel mechanisms of pain in sickle cell disease. Blood. 2017;130(22):2377-2385. doi:10.1182/blood-2017-05-782003
    1. Kohli DR, Li Y, Khasabov SG, et al. . Pain-related behaviors and neurochemical alterations in mice expressing sickle hemoglobin: modulation by cannabinoids. Blood. 2010;116(3):456-465. doi:10.1182/blood-2010-01-260372
    1. Vincent L, Vang D, Nguyen J, Benson B, Lei J, Gupta K. Cannabinoid receptor-specific mechanisms to alleviate pain in sickle cell anemia via inhibition of mast cell activation and neurogenic inflammation. Haematologica. 2016;101(5):566-577. doi:10.3324/haematol.2015.136523
    1. Cain DM, Vang D, Simone DA, Hebbel RP, Gupta K. Mouse models for studying pain in sickle disease: effects of strain, age, and acuteness. Br J Haematol. 2012;156(4):535-544. doi:10.1111/j.1365-2141.2011.08977.x
    1. Elikkottil J, Gupta P, Gupta K. The analgesic potential of cannabinoids. J Opioid Manag. 2009;5(6):341-357. doi:10.5055/jom.2009.0034
    1. Abrams DI, Jay CA, Shade SB, et al. . Cannabis in painful HIV-associated sensory neuropathy: a randomized placebo-controlled trial. Neurology. 2007;68(7):515-521. doi:10.1212/01.wnl.0000253187.66183.9c
    1. Andreae MH, Carter GM, Shaparin N, et al. . Inhaled cannabis for chronic neuropathic pain: a meta-analysis of individual patient data. J Pain. 2015;16(12):1221-1232. doi:10.1016/j.jpain.2015.07.009
    1. Mechoulam R, Parker LA, Gallily R. Cannabidiol: an overview of some pharmacological aspects. J Clin Pharmacol. 2002;42(S1):11S-19S. doi:10.1002/j.1552-4604.2002.tb05998.x
    1. Fasinu PS, Phillips S, ElSohly MA, Walker LA. Current status and prospects for cannabidiol preparations as new therapeutic agents. Pharmacotherapy. 2016;36(7):781-796. doi:10.1002/phar.1780
    1. Abrams DI, Vizoso HP, Shade SB, Jay C, Kelly ME, Benowitz NL. Vaporization as a smokeless cannabis delivery system: a pilot study. Clin Pharmacol Ther. 2007;82(5):572-578. doi:10.1038/sj.clpt.6100200
    1. Foltin RW, Fischman MW, Byrne MF. Effects of smoked marijuana on food intake and body weight of humans living in a residential laboratory. Appetite. 1988;11(1):1-14. doi:10.1016/S0195-6663(88)80017-5
    1. Tan G, Jensen MP, Thornby JI, Shanti BF. Validation of the Brief Pain Inventory for chronic nonmalignant pain. J Pain. 2004;5(2):133-137. doi:10.1016/j.jpain.2003.12.005
    1. Centers for Disease Control and Prevention CDC, FDA, states continue to investigate severe pulmonary disease among people who use e-cigarettes. Accessed June 15, 2020.
    1. Arepally GM, Ortel TL. Bad weed: synthetic cannabinoid-associated coagulopathy. Blood. 2019;133(9):902-905. doi:10.1182/blood-2018-11-876839
    1. Knight-Madden J, Lewis N, Hambleton IR. The prevalence of marijuana smoking in young adults with sickle cell disease: a longitudinal study. West Indian Med J. 2006;55(4):224-227. doi:10.1590/S0043-31442006000400004
    1. Howard J, Anie KA, Holdcroft A, Korn S, Davies SC. Cannabis use in sickle cell disease: a questionnaire study. Br J Haematol. 2005;131(1):123-128. doi:10.1111/j.1365-2141.2005.05723.x
    1. Ballas SK. The use of cannabis by patients with sickle cell disease increased the frequency of hospitalization due to vaso-occlusive crises. Cannabis Cannabinoid Res. 2017;2(1):197-201. doi:10.1089/can.2017.0011
    1. Roberts JD, Spodick J, Cole J, Bozzo J, Curtis S, Forray A. Marijuana use in adults living with sickle cell disease. Cannabis Cannabinoid Res. 2018;3(1):162-165. doi:10.1089/can.2018.0001
    1. Abrams DI, Couey P, Shade SB, Kelly ME, Benowitz NL. Cannabinoid-opioid interaction in chronic pain. Clin Pharmacol Ther. 2011;90(6):844-851. doi:10.1038/clpt.2011.188
    1. Karniol IG, Shirakawa I, Kasinski N, Pfeferman A, Carlini EA. Cannabidiol interferes with the effects of delta 9-tetrahydrocannabinol in man. Eur J Pharmacol. 1974;28(1):172-177. doi:10.1016/0014-2999(74)90129-0
    1. Solowij N, Broyd S, Greenwood LM, et al. . A randomised controlled trial of vaporised Δ9-tetrahydrocannabinol and cannabidiol alone and in combination in frequent and infrequent cannabis users: acute intoxication effects. Eur Arch Psychiatry Clin Neurosci. 2019;269(1):17-35. doi:10.1007/s00406-019-00978-2
    1. Bachhuber MA, Saloner B, Cunningham CO, Barry CL. Medical cannabis laws and opioid analgesic overdose mortality in the United States, 1999-2010. JAMA Intern Med. 2014;174(10):1668-1673. doi:10.1001/jamainternmed.2014.4005
    1. Bradford AC, Bradford WD, Abraham A, Bagwell Adams G. Association between US state medical cannabis laws and opioid prescribing in the Medicare Part D population. JAMA Intern Med. 2018;178(5):667-672. doi:10.1001/jamainternmed.2018.0266

Source: PubMed

3
購読する