Skip to main content
Log in

Critical Aspects Affecting Cannabidiol Oral Bioavailability and Metabolic Elimination, and Related Clinical Implications

  • Leading Article
  • Published:
CNS Drugs Aims and scope Submit manuscript

Abstract

This article provides a critical appraisal of the available evidence concerning clinical exposure to orally administered cannabidiol (CBD), with special reference to factors affecting gastrointestinal absorption, presystemic elimination, and susceptibility to metabolic drug interactions. Although detailed studies have not been published, the available data suggest that the absolute bioavailability of CBD after oral dosing under fasting conditions is approximately 6%, and increases fourfold when the medication is co-administered with a high-fat meal. Based on measurements of CBD plasma exposure after oral dosing and a 6% absolute oral bioavailability estimate, the actual clearance of CBD in adults can be inferred to be in the order of 67 L/h, which is similar to the value of 74 ± 14 L/h (mean ± standard deviation) determined after intravenous injection of a 20-mg dose of deuterium-labeled CBD in five healthy subjects. Assuming that the CBD blood-to-plasma ratio is about 1, as in the case of tetrahydrocannabinol (THC), and that CBD metabolism takes place virtually entirely in the liver, it can be estimated that about 70 to 75% of an orally absorbed dose of CBD can be removed by hepatic metabolism before reaching the systemic circulation, and additionally CBD gastrointestinal absorption is incomplete. A formulation with improved biopharmaceutical properties could increase the extent of CBD absorption about fourfold (i.e., to the level achieved with the currently available formulations co-administered with a high-fat meal) and minimize the influence of food effects on CBD bioavailability. There is also potential for favoring the absorption of CBD through the enteric lymphatic system, thereby reducing the extent of presystemic hepatic elimination. Evidence that CBD can behave as a high hepatic clearance compound also has implications when predicting the magnitude of drug–drug interactions affecting CBD metabolism. These considerations have important clinical relevance, particularly with respect to the objective of minimizing pharmacokinetic variability and consequent intra- and interindividual differences in therapeutic response and susceptibility to adverse effects.

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

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Similar content being viewed by others

References

  1. Amin MR, Ali DW. Pharmacology of medical cannabis. Adv Exp Med Biol. 2019;1162:151–65.

    Article  CAS  Google Scholar 

  2. Campos AC, Fogaça MV, Sonego AB, Guimarães FS. Cannabidiol, neuroprotection and neuropsychiatric disorders. Pharmacol Res. 2016;112:119–27.

    Article  CAS  Google Scholar 

  3. Elsaid S, Kloiber S, Le Foll B. Effects of cannabidiol (CBD) in neuropsychiatric disorders: a review of pre-clinical and clinical findings. Prog Mol Biol Transl Sci. 2019;167:25–75.

    Article  Google Scholar 

  4. Cassano T, Villani R, Pace L, Carbone A, Naik Bukke V, Orkisz S, et al. From Cannabis sativa to cannabidiol: promising therapeutic candidate for the treatment of neurodegenerative diseases. Front Pharmacol. 2020;11:124.

    Article  Google Scholar 

  5. Kis B, Ifrim FC, Buda V, Avram S, Pavel IZ, Antal D, et al. Cannabidiol-from plant to human body: a promising bioactive molecule with multi-target effects in cancer. Int J Mol Sci. 2019;20(23):E5905.

    Article  Google Scholar 

  6. Pisanti S, Malfitano AM, Ciaglia E, Lamberti A, Ranieri R, Cuomo G, et al. Cannabidiol: state of the art and new challenges for therapeutic applications. Pharmacol Ther. 2017;175:133–50.

    Article  CAS  Google Scholar 

  7. Fitzcharles MA, Clauw DJ, Hauser W. A cautious hope for cannabidiol (CBD) in rheumatology care. Arthritis Care Res. 2020. https://doi.org/10.1002/acr.24176.

    Article  Google Scholar 

  8. Franco V, Perucca E. Pharmacological and therapeutic properties of cannabidiol for epilepsy. Drugs. 2019;79:1435–54.

    Article  CAS  Google Scholar 

  9. Epidiolex. Full Prescribing Information. Greenwich Biosciences Inc. 2018. https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/210365lbl.pdf. Accessed 10 Apr 2020.

  10. Epidyolex. Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/product-information/epidyolex-epar-product-information_en.pdf. Accessed 10 Apr 2020.

  11. Geffrey AL, Pollack SF, Bruno PL, Thiele EA. Drug–drug interaction between clobazam and cannabidiol in children with refractory epilepsy. Epilepsia. 2015;56:1246–51.

    Article  CAS  Google Scholar 

  12. Morrison G, Crockett J, Blakey G, Sommerville K. A Phase 1, open-label, pharmacokinetic trial to investigate possible drug-drug interactions between clobazam, stiripentol, or valproate and cannabidiol in healthy subjects. Clin Pharmacol Drug Dev. 2019;8:1009–31.

    Article  CAS  Google Scholar 

  13. European Medicines Agency, Committee for Medicinal Products for Human Use (CHMP), Epidyolex Assessment Report, https://www.ema.europa.eu/en/documents/assessment-report/epidyolex-epar-public-assessment-report_en.pdf. Accessed 18 Mar 2020.

  14. Amidon GL, Lennernas H, Shah VP, Crison JR. A theoretical basis for biopharmaceutics drug classification: the correlation of in vitro drug product dissolution and in vivo bioavailability. Pharm Res. 1995;12:413–20.

    Article  CAS  Google Scholar 

  15. Wu C-Y, Benet LZ. Predicting drug disposition via application of BCS: transport/absorption/elimination interplay and development of a biopharmaceutics drug disposition classification system. Pharm Res. 2005;22:11–23.

    Article  CAS  Google Scholar 

  16. Chan R, Wei C-Y, Chen Y-T, Benet LZ. Use of the biopharmaceutics drug disposition classification system (BDDCS) to help predict the occurrence of idiosyncratic coetaneous adverse reaction associated with antiepileptic drug usage. AAPS J. 2016;18:757–76.

    Article  CAS  Google Scholar 

  17. DrugBank Database: Cannabidiol. https://www.drugbank.ca/drugs/DB09061. Accessed 24 Mar 2020.

  18. Harvey DJ, Samara E, Mechoulam R. Comparative metabolism of cannabidiol in dog, rat and man. Pharmacol Biochem Behav. 1991;40:523–32.

    Article  CAS  Google Scholar 

  19. Samara E, Bialer M, Harvey DJ. Pharmacokinetics of urinary metabolites of cannabidiol in the dog. Biopharm Drug Dispos. 1990;11:785–95.

    Article  CAS  Google Scholar 

  20. Gaston TE, Friedman D. Pharmacology of cannabinoids in the treatment of epilepsy. Epilepsy Behav. 2017;70(Pt B):313–8.

    Article  Google Scholar 

  21. Zendulka O, Dovrtelova G, Noskova K, Turjap M, Sulcova A, Hanus L, et al. Cannabinoids and cytochrome P450 interactions. Curr Drug Metab. 2016;17:206–26.

    Article  CAS  Google Scholar 

  22. Ujváry I, Hanuš L. Human metabolites of cannabidiol: a review on their formation, biological activity, and relevance in therapy. Cannabis Cannabinoid Res. 2016;1:90–101.

    Article  Google Scholar 

  23. Miller SA, Stone NL, Yates SAS, O’Sullivan SE. A systemic review on the pharmacokinetics of cannabidiol in humans. Front Pharmacol. 2018. https://doi.org/10.3389/fphar.2018.01365.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Bialer M, Johannessen SI, Koepp MJ, Levy RH, Perucca E, Tomson T, et al. Progress report on new antiepileptic drugs: a summary of the fourteen Eilat Conference on new antiepileptic drugs and devices. II. Drugs in more advanced clinical development. Epilepsia. 2018;59:1856–84.

    Google Scholar 

  25. Ohlsson A, Lindgren JE, Wahlen A, Agurell S, Hollister LE, Gillespie HK. Plasma delta-9-tetrahydrocannabinol concentrations and clinical effects after oral and intravenous administration and smoking. Clin Pharmacol Ther. 1980;28:409–16.

    Article  CAS  Google Scholar 

  26. Grotenhermen F. Pharmacokinetics and pharmacodynamics of cannabinoids. Clin Pharmacokinet. 2003;42:327–60.

    Article  CAS  Google Scholar 

  27. Ohlsson A, Lindgren JE, Andersson S, Agurell S, Gillespie H, Hollister LE. Single-dose kinetics of deuterium-labelled cannabidiol in man after smoking and intravenous administration. Biomed Environ Mass Spectrom. 1986;13:77–83.

    Article  CAS  Google Scholar 

  28. Taylor L, Gidal B, Blakey G, Tayo B, Morrison G. A phase I, randomized, double-blind, placebo-controlled, single ascending dose, multiple dose, and food effect trial of the safety, tolerability and pharmacokinetics of highly purified cannabidiol in healthy subjects. CNS Drugs. 2018;32:1053–67.

    Article  CAS  Google Scholar 

  29. Birnbaum AK, Karanam A, Marino SE, Barkley CM, Remmel RP, Roslawski M, et al. Food effect on pharmacokinetics of cannabidiol oral capsules in adult patients with refractory epilepsy. Epilepsia. 2019;60:1586–92.

    Article  CAS  Google Scholar 

  30. Crockett J, Critchley D, Tayo B, Berwaerts J, Morrison G. A phase 1, randomized, pharmacokinetic trial of the effect of different meal compositions, whole milk, and alcohol on cannabidiol exposure and safety in healthy subjects. Epilepsia. 2020;61:267–77.

    Article  CAS  Google Scholar 

  31. Bialer M, Arcavi L, Sussan S, Volosov A, Yacobi D, Moros D, et al. Existing and new criteria for bioequivalent of new controlled release (CR) products of carbamazepine. Epilepsy Res. 1998;32:371–8.

    Article  CAS  Google Scholar 

  32. Taylor L, Crockett J, Tayo B, Morrison G. A Phase 1, open-label, parallel-group, single-dose trial of the pharmacokinetics and safety of cannabidiol (CBD) in subjects with mild to severe hepatic impairment. J Clin Pharmacol. 2019;59:1110–9.

    Article  CAS  Google Scholar 

  33. Tayo B, Taylor L, Sahebkar F, Morrison G. A Phase 1, open-label, parallel-group, single-dose trial of the pharmacokinetics, safety and tolerability of cannabidiol in subjects with mild to severe renal impairment. Clin Pharmacokinet. 2019. https://doi.org/10.1007/s40262-019-00841-6(Epub ahead of print).

    Article  PubMed Central  Google Scholar 

  34. Widman M, Agurell S, Ehrnebo M, Jones G. Binding of (+)- and (–)-Δ1-tetrahydrocannabinols and (–)-7-hydroxy-Δ1-tetrahydrocannabinol to blood cells and plasma proteins in man. J Pharm Pharmacol. 1974;26:914–6.

    Article  CAS  Google Scholar 

  35. Wilkinson GR, Shand DG. Commentary: a physiological approach to hepatic drug clearance. Clin Pharmacol Ther. 1975;18:377–90.

    Article  CAS  Google Scholar 

  36. Derendorf H, Schmidt S. Rowland and Tozer’s clinical pharmacokinetics and pharmacodynamics. 5th ed. South Holland: Wloters Kluwer; 2020. p. 78–82.

    Google Scholar 

  37. Klumpers LE, Beumer TL, van Hasselt JG, Lipplaa A, Karger LB, Kleinloog HD, et al. Novel delta(9)-tetrahydrocannabinol formulation Namisol has beneficial pharmacokinetics and promising pharmacodynamic effects. Br J Clin Pharmacol. 2012;74:42–53.

    Article  CAS  Google Scholar 

  38. Marinol. Full Precribing information. Patheon Softgels Inc. 2017. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/018651s029lbl.pdf. Accessed 11 Apr 2020.

  39. Zgair A, Wong JC, Lee JB, Mistry J, Sivak O, Wasan KM, et al. Dietary fats and pharmaceutical lipid excipients increase systemic exposure to orally administered cannabis and cannabis-based medicines. Am J Transl Res. 2016;8:3448–59.

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Lee JB, Zgair A, Malec J, Kim TH, Kim MG, et al. Lipophilic activated ester prodrug approach for drug delivery to the intestinal lymphatic system. J Control Release. 2018;286:10–9.

    Article  CAS  Google Scholar 

  41. Brocks DR, Davies NM. Lymphatic drug absorption via the enterocytes: pharmacokinetic simulation, modeling, and considerations for optimal dug development. J Pharm Pharm Sci. 2018;21:254s–70s.

    Article  Google Scholar 

  42. Whalley BJ, Stott C, Gray RA. The human metabolite of cannabidiol, 7-hydroxy-cannabidiol, but not 7-carboxy-cannabidiol, is anticonvulsant in the maximal electroshock threshold test (MEST) in mouse [abstract no. 1.435]. 71st Annual Meeting of the American Epilepsy Society, Washington DS, December 1–5, 2017. https://www.aesnet.org/meetings_events/annual_meeting_abstracts/view/381222. Accessed 11 Apr 2020.

  43. Jiang R, Yamaori S, Takeda S, Yamamoto I, Watanabe K. Identification of cytochrome P450 enzymes responsible for metabolism of cannabidiol by human liver microsomes. Life Sci. 2011;89:165–70.

    Article  CAS  Google Scholar 

  44. Stott C, White L, Wright S, Wilbraham D, Guy G. 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. 2013;2:236.

    Article  Google Scholar 

  45. Garnock-Jones KP. Oromucosal midazolam: a review of its use in pediatric patients with prolonged acute convulsive seizures. Paediatr Drugs. 2012;14:251–61.

    Article  Google Scholar 

  46. Tran A, Rey E, Pons G, Rousseau M, d’Athis P, Olive G, et al. Influence of stiripentol on cytochrome P450-mediated metabolic pathways in humans: in vitro and in vivo comparison and calculation of in vivo inhibition constants. Clin Pharmacol Ther. 1997;62:490–504.

    Article  CAS  Google Scholar 

  47. Arzimanoglou A, French J, Blume WT, Cross JH, Ernst JP, et al. Lennox–Gastaut syndrome: a consensus approach on diagnosis, assessment, management, and trial methodology. Lancet Neurol. 2009;8:82–93.

    Article  Google Scholar 

  48. Wirrell EC. Treatment of Dravet syndrome. Can J Neurol Sci. 2016;43(Suppl 3):S13–8.

    Article  Google Scholar 

  49. Thiele E, Bebin EM, Bhathal H, Jansen F, Kotulska-Jozwiak K, et al. Cannabidiol (CBD) treatment in patients with seizures associated with tuberous sclerosis complex: a randomized double-blind, placebo-controlled Phase 3 trial (GWPCARE6). Abstract. Annual Meeting of the American Epilepsy Society, Baltimore, Maryland, December 6–10, 2019. https://www.aesnet.org/meetings_events/annual_meeting_abstracts/view/2421288. Accessed 10 Apr 2020.

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Meir Bialer.

Ethics declarations

Funding

This work was not supported by any funding source.

Conflict of interest

MB has received speaker’s or consultancy fees from Alkaloid, Boehringer Ingelheim, Medison, and US WorldMeds. EP has received speaker’s or consultancy fees from Amicus Therapeutics, Arvelle, Biogen, Eisai, GW Pharma, Intas Pharmaceuticals, Laboratorios Bagò, Sanofi, Sun Pharma, UCB Pharma, and Xenon Pharma.

Ethical approval

We confirm that we have read the journal’s position on issues involved in ethical publication and affirm that this report is consistent with those guidelines.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Perucca, E., Bialer, M. Critical Aspects Affecting Cannabidiol Oral Bioavailability and Metabolic Elimination, and Related Clinical Implications. CNS Drugs 34, 795–800 (2020). https://doi.org/10.1007/s40263-020-00741-5

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40263-020-00741-5

Navigation