A Systematic Review on the Pharmacokinetics of Cannabidiol in Humans

Sophie A Millar, Nicole L Stone, Andrew S Yates, Saoirse E O'Sullivan, Sophie A Millar, Nicole L Stone, Andrew S Yates, Saoirse E O'Sullivan

Abstract

Background: Cannabidiol is being pursued as a therapeutic treatment for multiple conditions, usually by oral delivery. Animal studies suggest oral bioavailability is low, but literature in humans is not sufficient. The aim of this review was to collate published data in this area. Methods: A systematic search of PubMed and EMBASE (including MEDLINE) was conducted to retrieve all articles reporting pharmacokinetic data of CBD in humans. Results: Of 792 articles retireved, 24 included pharmacokinetic parameters in humans. The half-life of cannabidiol was reported between 1.4 and 10.9 h after oromucosal spray, 2-5 days after chronic oral administration, 24 h after i.v., and 31 h after smoking. Bioavailability following smoking was 31% however no other studies attempted to report the absolute bioavailability of CBD following other routes in humans, despite i.v formulations being available. The area-under-the-curve and Cmax increase in dose-dependent manners and are reached quicker following smoking/inhalation compared to oral/oromucosal routes. Cmax is increased during fed states and in lipid formulations. Tmax is reached between 0 and 4 h. Conclusions: This review highlights the paucity in data and some discrepancy in the pharmacokinetics of cannabidiol, despite its widespread use in humans. Analysis and understanding of properties such as bioavailability and half-life is critical to future therapeutic success, and robust data from a variety of formulations is required.

Keywords: CMAX; TMAX; bioavailability; endocannabinoid system; half life; pharmacokinetics; plasma clearance; volume of distribution.

Figures

Figure 1
Figure 1
Flow chart for study retrieval and selection.
Figure 2
Figure 2
(A) Mean or median Tmax (h) and range against CBD dose (mg) (B) mean or median area under the curve (AUC0-t) (h × ng/mL) and SD against CBD dose (mg) and (C) plasma mean or median concentration max (Cmax; ng/mL) against CBD dose (mg). It was not possible to present error bars for Cmax as SD and SEM were both reported in the data. IV, intravenous; SD, standard deviation; SEM, standard error of the mean.

References

    1. Al Saabi A., Allorge D., Sauvage F. L., Tournel G., Gaulier J. M., Marquet P., et al. . (2013). Involvement of UDP-glucuronosyltransferases UGT1A9 and UGT2B7 in ethanol glucuronidation, and interactions with common drugs of abuse. Drug Metab. Dispos. 41, 568–574. 10.1124/dmd.112.047878
    1. Atsmon J., Cherniakov I., Izgelov D., Hoffman A., Domb A. J., Deutsch L., et al. . (2017a). PTL401, a new formulation based on pro-nano dispersion technology, improves oral cannabinoids bioavailability in healthy volunteers. J. Pharm. Sci. 107, 1423–1429. 10.1016/j.xphs.2017.12.020
    1. Atsmon J., Heffetz D., Deutsch L., Deutsch F., Sacks H. (2017b). Single-Dose pharmacokinetics of oral cannabidiol following administration of PTL101: a new formulation based on gelatin matrix pellets technology. Clin. Pharmacol. Drug Dev. 7:751–758. 10.1002/cpdd.408
    1. Bergamaschi M. M., Queiroz R. H., Zuardi A. W., Crippa J. A. (2011). Safety and side effects of cannabidiol, a Cannabis sativa constituent. Curr. Drug Saf. 6, 237–249. 10.2174/157488611798280924
    1. Chagas M. H., Zuardi A. W., Tumas V., Pena-Pereira M. A., Sobreira E. T., Bergamaschi M. M., et al. . (2014). Effects of cannabidiol in the treatment of patients with Parkinson's disease: an exploratory double-blind trial. J. Psychopharmacol. 28, 1088–1098. 10.1177/0269881114550355
    1. Cherniakov I., Izgelov D., Barasch D., Davidson E., Domb A. J., Hoffman A. (2017a). Piperine-pro-nanolipospheres as a novel oral delivery system of cannabinoids: pharmacokinetic evaluation in healthy volunteers in comparison to buccal spray administration. J. Control. Release 266, 1–7. 10.1016/j.jconrel.2017.09.011
    1. Cherniakov I., Izgelov D., Domb A. J., Hoffman A. (2017b). The effect of Pro NanoLipospheres (PNL) formulation containing natural absorption enhancers on the oral bioavailability of delta-9-tetrahydrocannabinol (THC) and cannabidiol (CBD) in a rat model. Eur. J. Pharm. Sci. 109, 21–30. 10.1016/j.ejps.2017.07.003
    1. Consroe P., Kennedy K., Schram K. (1991). Assay of plasma cannabidiol by capillary gas chromatography/ion trap mass spectroscopy following high-dose repeated daily oral administration in humans. Pharmacol. Biochem. Behav. 40, 517–522. 10.1016/0091-3057(91)90357-8
    1. Crippa J. A., Derenusson G. N., Ferrari T. B., Wichert-Ana L., Duran F. L., Martin-Santos R., et al. . (2011). Neural basis of anxiolytic effects of cannabidiol (CBD) in generalized social anxiety disorder: a preliminary report. J. Psychopharmacol. 25, 121–130. 10.1177/0269881110379283
    1. Deiana S., Watanabe A., Yamasaki Y., Amada N., Arthur M., Fleming S., et al. (2012)Plasma brain pharmacokinetic profile of cannabidiol (CBD), cannabidivarine (CBDV), Delta(9)-tetrahydrocannabivarin (THCV) cannabigerol (CBG) in rats mice following oral intraperitoneal administration CBD action on obsessive-compulsive behaviour. Psychopharmacology 219, 859–873. 10.1007/s00213-011-2415-0.
    1. Desrosiers N. A., Himes S. K., Scheidweiler K. B., Concheiro-Guisan M., Gorelick D. A., Huestis M. A. (2014). Phase I and II cannabinoid disposition in blood and plasma of occasional and frequent smokers following controlled smoked cannabis. Clin. Chem. 60, 631–643. 10.1373/clinchem.2013.216507
    1. Devinsky O., Cross J. H., Wright S. (2017). Trial of Cannabidiol for drug-resistant seizures in the Dravet Syndrome. N. Engl. J. Med. 377, 699–700. 10.1056/NEJMc1708349
    1. Devinsky O., Patel A. D., Cross J. H., Villanueva V., Wirrell E. C., Privitera M., et al. . (2018a). Effect of Cannabidiol on drop seizures in the Lennox-Gastaut Syndrome. N. Engl. J. Med. 378, 1888–1897. 10.1056/NEJMoa1714631
    1. Devinsky O., Patel A. D., Thiele E. A., Wong M. H., Appleton R., Harden C. L., et al. . (2018b). Randomized, dose-ranging safety trial of cannabidiol in Dravet syndrome. Neurology 90, e1204–e1211. 10.1212/WNL.0000000000005254
    1. Eichler M., Spinedi L., Unfer-Grauwiler S., Bodmer M., Surber C., Luedi M., et al. . (2012). Heat exposure of Cannabis sativa extracts affects the pharmacokinetic and metabolic profile in healthy male subjects. Planta Med. 78, 686–691. 10.1055/s-0031-1298334
    1. Fattore L., Fratta W. (2010). How important are sex differences in cannabinoid action? Br. J. Pharmacol. 160, 544–548. 10.1111/j.1476-5381.2010.00776.x
    1. Fernandez E., Perez R., Hernandez A., Tejada P., Arteta M., Ramos J. T. (2011). Factors and mechanisms for pharmacokinetic differences between pediatric population and adults. Pharmaceutics 3, 53–72. 10.3390/pharmaceutics3010053
    1. Garberg H. T., Solberg R., Barlinn J., Martinez-Orgado J., Loberg E. M., Saugstad O. D. (2017). High-dose cannabidiol induced hypotension after global hypoxia-ischemia in piglets. Neonatology 112, 143–149. 10.1159/000471786
    1. Gaston T. E., Bebin E. M., Cutter G. R., Liu Y., Szaflarski J. P. (2017). Interactions between cannabidiol and commonly used antiepileptic drugs. Epilepsia 58, 1586–1592. 10.1111/epi.13852
    1. Geffrey A. L., Pollack S. F., Bruno P. L., Thiele E. A. (2015). Drug-drug interaction between clobazam and cannabidiol in children with refractory epilepsy. Epilepsia 56, 1246–1251. 10.1111/epi.13060
    1. Giacoppo S., Galuppo M., Pollastro F., Grassi G., Bramanti P., Mazzon E. (2015). A new formulation of cannabidiol in cream shows therapeutic effects in a mouse model of experimental autoimmune encephalomyelitis. Daru 23:48. 10.1186/s40199-015-0131-8
    1. Gunasekaran N., Long L., Dawson B., Hansen G., Richardson D., Li K., et al. (2009). Reintoxication: the release of fat-stored Δ9-tetrahydrocannabinol (THC) into blood is enhanced by food deprivation or ACTH exposure. Br. J. Pharmacol. 158, 1330–1337. 10.1111/j.1476-5381.2009.00399.x
    1. Guy G. W., Flint M. E. (2004). A single centre, placebo-controlled, four period, crossover, tolerability study assessing, pharmacodynamic effects, pharmacokinetic characteristics and cognitive profiles of a single dose of three formulations of Cannabis Based Medicine Extracts (CBMEs) (GWPD9901), plus a two period tolerability study comparing pharmacodynamic effects and pharmacokinetic characteristics of a single dose of a cannabis based medicine extract given via two administration routes (GWPD9901 EXT). J. Cannabis Ther. 3, 35–77. 10.1300/J175v03n03_03
    1. Guy G. W., Robson P. J. (2004a). A phase I, double blind, three-way crossover study to assess the pharmacokinetic profile of Cannabis Based Medicine Extract (CBME) administered sublingually in variant cannabinoid ratios in normal healthy male volunteers. J. Cannabis Ther. 3, 121–152. 10.1300/J175v03n04_02
    1. Guy G. W., Robson P. J. (2004b). A phase I, open label, four-way crossover study to compare the pharmacokinetic profiles of a single dose of 20 mg of a Cannabis Based Medicine Extract (CBME) Administered on 3 different areas of the buccal mucosa and to investigate the pharmacokinetics of CBME per Oral in Healthy Male and Female Volunteers (GWPK0112). J. Cannabis Ther. 3, 79–120. 10.1300/J175v03n04_01
    1. GW Pharmaceuticals (2018). Subsidiary Greenwich Biosciences Announce FDA Approval of EPIDIOLEX® (Cannabidiol) Oral Solution–The First Plant-Derived Cannabinoid Prescription Medicine.
    1. Hammell D. C., Zhang L. P., Ma F., Abshire S. M., McIlwrath S. L., Stinchcomb A. L., et al. . (2016). Transdermal cannabidiol reduces inflammation and pain-related behaviours in a rat model of arthritis. Eur. J. Pain 20, 936–948. 10.1002/ejp.818
    1. Haney M., Malcolm R. J., Babalonis S., Nuzzo P. A., Cooper Z. D., Bedi G., et al. . (2016). Oral cannabidiol does not alter the subjective, reinforcing or cardiovascular effects of smoked cannabis. Neuropsychopharmacology 41, 1974–1982. 10.1038/npp.2015.367
    1. Hemp Business (2017). Hemp Business Journal. State of Hemp Market Report.
    1. Huestis M. A. (2007). Human cannabinoid pharmacokinetics. Chem. Biodivers. 4, 1770–1804. 10.1002/cbdv.200790152
    1. Izzo A. A., Borrelli F., Capasso R., Di Marzo V., Mechoulam R. (2009). Non-psychotropic plant cannabinoids: new therapeutic opportunities from an ancient herb. Trends Pharmacol. Sci. 30, 515–527. 10.1016/j.tips.2009.07.006
    1. Jiang R., Yamaori S., Okamoto Y., Yamamoto I., Watanabe K. (2013). Cannabidiol is a potent inhibitor of the catalytic activity of cytochrome P450 2C19. Drug Metab. Pharmacokinet. 28, 332–338. 10.2133/dmpk.DMPK-12-RG-129
    1. Karschner E. L., Darwin W. D., Goodwin R. S., Wright S., Huestis M. A. (2011). Plasma cannabinoid pharmacokinetics following controlled oral Δ9-tetrahydrocannabinol and oromucosal cannabis extract administration. Clin. Chem. 57, 66–75. 10.1373/clinchem.2010.152439
    1. Lee D., Schwope D. M., Milman G., Barnes A. J., Gorelick D. A., Huestis M. A. (2012). Cannabinoid disposition in oral fluid after controlled smoked cannabis. Clin. Chem. 58, 748–756. 10.1373/clinchem.2011.177881
    1. Leweke F. M., Piomelli D., Pahlisch F., Muhl D., Gerth C. W., Hoyer C., et al. . (2012). Cannabidiol enhances anandamide signaling and alleviates psychotic symptoms of schizophrenia. Transl. Psychiatry 2:e94. 10.1038/tp.2012.15
    1. Long L. E., Chesworth R., Huang X. F., Wong A., Spiro A., McGregor I. S., et al. . (2012). Distinct neurobehavioural effects of cannabidiol in transmembrane domain neuregulin 1 mutant mice. PLoS ONE 7:e34129. 10.1371/journal.pone.0034129
    1. Manini A. F., Yiannoulos G., Bergamaschi M. M., Hernandez S., Olmedo R., Barnes A. J., et al. . (2015). Safety and pharmacokinetics of oral cannabidiol when administered concomitantly with intravenous fentanyl in humans. J. Addict. Med. 9, 204–210. 10.1097/ADM.0000000000000118
    1. Mechoulam R., Parker L. A., Gallily R. (2002). Cannabidiol: an overview of some pharmacological aspects. J. Clin. Pharmacol. 42, 11S−19S. 10.1002/j.1552-4604.2002.tb05998.x
    1. Moher D., Liberati A., Tetzlaff J., Altman D. G. (2009). Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. Ann. Int. Med. 151, 264–269, w64. 10.7326/0003-4819-151-4-200908180-00135
    1. Nadulski T., Pragst F., Weinberg G., Roser P., Schnelle M., Schnelle M., Fronk E. M., et al. (2005a). Randomized, double-blind, placebo-controlled study about the effects of Cannabidiol (CBD) on the pharmacokinetics of D9-Tetrahydrocannabinol (THC) after oral application of THC verses standardized cannabis extract. Ther. Drug Monit. 27, 799–810. 10.1097/01.ftd.0000177223.19294.5c
    1. Nadulski T., Sporkert F., Schnelle M., Stadelmann A. M., Roser P., Schefter T., et al. . (2005b). Simultaneous and sensitive analysis of THC, 11-OH-THC, THC-COOH, CBD, and CBN by GC-MS in plasma after oral application of small doses of THC and cannabis extract. J. Anal. Toxicol. 29, 782–789. 10.1093/jat/29.8.782
    1. Naftali T., Mechulam R., Marii A., Gabay G., Stein A., Bronshtain M., et al. (2017). Low-dose cannabidiol is safe but not effective in the treatment for crohn's disease, a randomized controlled trial. Dig. Dis. Sci. 62, 1615–1620. 10.1007/s10620-017-4540-z
    1. National Institute for Health (2014). National Lung and Blood Institute Quality Assessment Tool for Before-After (Pre-Post) Studies with No Control Group. Available online at:
    1. Newmeyer M. N., Desrosiers N. A., Lee D., Mendu D. R., Barnes A. J., Gorelick D. A., et al. . (2014). Cannabinoid disposition in oral fluid after controlled cannabis smoking in frequent and occasional smokers. Drug Test. Anal. 6, 1002–1010. 10.1002/dta.1632
    1. Ogungbenro K., Aarons L., Graham G. (2006). Sample size calculations based on generalized estimating equations for population pharmacokinetic experiments. J. Biopharm. Stat. 16, 135–150. 10.1080/10543400500508705
    1. Ohlsson A., Lindgren J. E., Andersson S., Agurell S., Gillespie H., Hollister L. E. (1986). Single-dose kinetics of deuterium-labelled cannabidiol in man after smoking and intravenous administration. Biomed. Environ. Mass Spectrom. 13, 77–83. 10.1002/bms.1200130206
    1. Schwope D. M., Karschner E. L., Gorelick D. A., Huestis M. A. (2011). Identification of recent cannabis use: whole-blood and plasma free and glucuronidated cannabinoid pharmacokinetics following controlled smoked cannabis administration. Clin. Chem. 57, 1406–1414. 10.1373/clinchem.2011.171777
    1. Sellers E. M., Schoedel K., Bartlett C., Romach M., Russo E. B., Stott C. G., et al. . (2013). A multiple-dose, randomized, double-blind, placebo-controlled, parallel-group QT/QTc study to evaluate the electrophysiologic effects of THC/CBD spray. Clin. Pharmacol. Drug Dev. 2, 285–294. 10.1002/cpdd.36
    1. Stott C., White L., Wright S., Wilbraham D., Guy G. (2013c). A Phase I, open-label, randomized, crossover study in three parallel groups to evaluate the effect of Rifampicin, Ketoconazole, and Omeprazole on the pharmacokinetics of THC/CBD oromucosal spray in healthy volunteers. Springerplus 2:236. 10.1186/2193-1801-2-236
    1. Stott C. G., White L., Wright S., Wilbraham D., Guy G. W. (2013a). A phase I study to assess the single and multiple dose pharmacokinetics of THC/CBD oromucosal spray. Eur. J. Clin. Pharmacol. 69, 1135–1147. 10.1007/s00228-012-1441-0
    1. Stott C. G., White L., Wright S., Wilbraham D., Guy G. W. (2013b). A phase I study to assess the effect of food on the single dose bioavailability of the THC/CBD oromucosal spray. Eur. J. Clin. Pharmacol. 69, 825–834. 10.1007/s00228-012-1393-4
    1. Stout S. M., Cimino N. M. (2014). Exogenous cannabinoids as substrates, inhibitors, and inducers of human drug metabolizing enzymes: a systematic review. Drug Metab. Rev. 46, 86–95. 10.3109/03602532.2013.849268
    1. Swortwood M. J., Newmeyer M. N., Andersson M., Abulseoud O. A., Scheidweiler K. B., Huestis M. A. (2017). Cannabinoid disposition in oral fluid after controlled smoked, vaporized, and oral cannabis administration. Drug Test. Anal. 9, 905–915. 10.1002/dta.2092
    1. Thiele E. A., Marsh E. D., French J. A., Mazurkiewicz-Beldzinska M., Benbadis S. R., Joshi C., et al. . (2018). Cannabidiol in patients with seizures associated with Lennox-Gastaut syndrome (GWPCARE4): a randomised, double-blind, placebo-controlled phase 3 trial. Lancet 391, 1085–1096. 10.1016/S0140-6736(18)30136-3
    1. Winter H., Ginsberg A., Egizi E., Erondu N., Whitney K., Pauli E., et al. . (2013). Effect of a high-calorie, high-fat meal on the bioavailability and pharmacokinetics of PA-824 in healthy adult subjects. Antimicrob. Agents Chemother. 57, 5516–5520. 10.1128/AAC.00798-13
    1. Zgair A., Wong J. C., Lee J. B., Mistry J., Sivak O., Wasan K. M., et al. . (2016). Dietary fats and pharmaceutical lipid excipients increase systemic exposure to orally administered cannabis and cannabis-based medicines. Am. J. Transl. Res. 8, 3448–3459.

Source: PubMed

3
S'abonner