Skip to main content
Log in

Minimal Physiologically Based Pharmacokinetic Model of Intravenously and Orally Administered Delta-9-Tetrahydrocannabinol in Healthy Volunteers

  • Original Research Article
  • Published:
European Journal of Drug Metabolism and Pharmacokinetics Aims and scope Submit manuscript

Abstract

Background and Objectives

Lack of information on the pharmacokinetics of the active moiety of Cannabis or the metabolites of delta-9-tetrahydrocannabinol (THC) does not seem to be discouraging medical or recreational use. Cytochrome P450 (CYP) 2C9, the primary enzyme responsible for THC metabolism, has two single nucleotide polymorphisms—Arg144Cys (*2) and Ile359Leu (*3). In the Caucasian population, allelic frequency is between 0.08 and 0.14 for CYP2C9*2 and between 0.04 and 0.16 for CYP2C9*3. In vitro data suggest that metabolic capacity for the variants CYP2C9*2 and CYP2C9*3 is about one-third compared to wild-type CYP2C9. Previous work has suggested exposure to the terminal metabolite is genetically determined. We therefore sought to characterize the pharmacokinetics of THC and its major metabolites 11-hydroxy-delta-9-tetrahydrocannabinol (THC-OH) and 11-nor-9-carboxy-delta-9-tetrahydrocannabinol (THC-COOH) in healthy volunteers with known CYP2C9 status by non-compartmental analysis (NCA), compartmental modeling (CM) and minimal physiologically based pharmacokinetic (mPBPK) modeling.

Methods

Blood samples drawn for THC, THC-OH and THC-COOH after a single intravenous (IV) bolus of 0.1 mg/kg (0.32 μM/kg) THC were analyzed using a validated LC–MS/MS method. NCA generated initial estimates and CM and the mPBPK model were then fit to plasma concentration data using non-linear mixed-effects modeling. Blood samples from orally dosed (10, 25 and 50 mg) THC brownies were added to validate the model.

Results

THC can be described as a high hepatic extraction ratio drug with blood flow-dependent metabolism not restricted by protein binding. THC hepatic clearance is dependent on the CYP2C9 genetic variant in the population. High extraction drugs display route-dependent metabolism. When administered via the IV or inhalation routes, induction or inhibition of CYP2C9 should be non-contributory as the elimination of THC is dependent only on liver blood flow. THC-OH is also a high extraction ratio drug, but its hepatic clearance is significantly impacted by the hepatic diffusional barrier that impedes its access to hepatic CYP2C9. THC-COOH is glucuronidated and renally cleared; subjects homozygous for CYP2C9*3 have reduced exposure to this metabolite as a result of the polymorphism reducing THC production, the hepatic diffusional barrier impeding egress from the hepatocyte, and increased renal clearance.

Conclusion

It has recently been reported that the terminal metabolite THC-COOH is active, implying the exposure difference in individuals homozygous for CYP2C9*3 may become therapeutically relevant. Defining the metabolism of THC in humans is important, as it is increasingly being used as a drug to treat various diseases and its recreational use is also rising. We have used NCA, CM, and mPBPK modeling of THC and its metabolites to partially disentangle the complexity of cannabis disposition in humans.

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.

Fig. 1
Fig. 2
Fig. 3

Similar content being viewed by others

References

  1. Legal Medical Marijuana States and DC. http://medicalmarijuana.procon.org. Accessed 28 Jan 2019.

  2. Huestis MA. Pharmacokinetics and metabolism of the plant cannabinoids, delta-9-tetrahydrocannabinol, cannabidiol and cannabinol. Handb Exp Pharmacol. 2005;168:657–90.

    Article  CAS  Google Scholar 

  3. Watanabe K, Yamaori S, Funahashi T, Kimura T, Yamamoto I. Cytochrome P450 enzymes involved in the metabolism of tetrahydrocannabinols and cannabinol by human hepatic microsomes. Life Sci. 2007;80(15):1415–9.

    Article  CAS  PubMed  Google Scholar 

  4. Bland TM, Haining RL, Tracy TS, Callery PS. CYP2C-catalyzed delta9-tetrahydrocannabinol metabolism: kinetics, pharmacogenetics and interaction with phenytoin. Biochem Pharmacol. 2005;70(7):1096–103.

    Article  CAS  PubMed  Google Scholar 

  5. Kirchheiner J, Brockmoller J. Clinical consequences of cytochrome P450 2C9 polymorphisms. Clin Pharmacol Ther. 2005;77(1):1–16.

    Article  CAS  PubMed  Google Scholar 

  6. Sachse-Seeboth C, Pfeil J, Sehrt D, et al. Interindividual variation in the pharmacokinetics of Delta9-tetrahydrocannabinol as related to genetic polymorphisms in CYP2C9. Clin Pharmacol Ther. 2009;85(3):273–6.

    Article  CAS  PubMed  Google Scholar 

  7. Kleine-Brueggeney M, Greif R, Brenneisen R, Urwyler N, Stueber F, Theiler LG. Intravenous delta-9-tetrahydrocannabinol to prevent postoperative nausea and vomiting: a randomized controlled trial. Anesth Analg. 2015;121(5):1157–64.

    Article  CAS  PubMed  Google Scholar 

  8. Vandrey R, Herrman ES, Mithcell JM, et al. Pharmacokinetic profile of oral cannabis in humans: blood and oral fluid disposition in relation to phamacodynamic outcomes. J Anal Toxicol. 2017;41:83–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Bernhard W, Wolowich WR, Sachs H, Kleine-Brueggeney M, Greif R, Theiler L. Influence of the genetic polymorphism in CYP2C9 on the pharmacokinetics of delta-9-tetrahydrocannabinol and introduction of a new pharmacokinetic model. In: Abstract K42. Proceedings of the American Academy of Forensic Sciences 66th annual meeting. Seattle, WA, February 17–22, 2014.

  10. Fiserova-Bergerova V. Inhalation anesthesia using physiologically based pharmacokinetic models. Drug Metab Rev. 1992;24:531–57.

    Article  CAS  PubMed  Google Scholar 

  11. Li GF, Shan ZQ. Modeling drug disposition and drug-drug interactions through hypothesis-driven physiologically based pharmacokinetics: a reversal translation perspective. Eur J Drug Metab Pharmacokinet. 2018;43:369–71.

    Article  CAS  PubMed  Google Scholar 

  12. Tsamandouras N, Rostami-Hodjegaan A, Aarons L. Combining the bottom up and top down approaches in pharmacokinetic modelling: fitting PBPK models to observed clinical data. Br J Clin Pharmacol. 2013;79:48–55.

    Article  CAS  PubMed Central  Google Scholar 

  13. Wishart DS, Feunang YD, Guo AC, Lo EJ, Marcu A, Grant JR, Sajed T, Johnson D, Li C, Sayeeda Z, Assempour N, Iynkkaran I, Liu Y, Maciejewski A, Gale N, Wilson A, Chin L, Cummings R, Le D, Pon A, Knox C, Wilson M. DrugBank 5.0: a major update to the DrugBank database for 2018. Nucleic Acids Res. 2017. https://doi.org/10.1093/nar/gkx1037.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Brown RP, Delp MD, Lindstedt SL, Rhomberg LR, Belilies RP. Physiological parameter values for physiologically based pharmacokinetic models. Toxicol Ind Health. 1997;13:407–84.

    Article  CAS  PubMed  Google Scholar 

  15. Rowland M, Tozer TN. Metabolite kinetics. In: Rowland M, Tozer TN, editors. Clinical pharmacokinetics: concepts and applications. 2nd ed. Malvern: Lea & Febiger; 1989.

    Google Scholar 

  16. Bornheim LM, Lasker JM, Raucy JL. Human hepatic microsomal metabolism of delta-1-tetrahydrocannabinol. Drug Metab Dispos. 1992;20:241–6.

    CAS  PubMed  Google Scholar 

  17. Mazur A, Lichti CF, Prather PL, et al. Characterization of human hepatic and extrahepatic UDP-glucuronosyltransferase enzymes involved in the metabolism of classic cannabinoids. Drug Metab Dispos. 2009;37:1496–504.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Dietz L, Glaz-Sandberg A, Nguyen H, Skopp G, Mikus G, Aderjan R. The urinary disposition of intravenously-administered 11-nor-9-carboxy-delta-9-tetrahydrocannabinol in humans. Ther Drug Monit. 2007;3:368–72.

    Article  CAS  Google Scholar 

  19. Cockcroft DW, Gault MH. Prediction of creatinine clearance from serum creatinine. Nephron. 1976;16:31–41.

    Article  CAS  PubMed  Google Scholar 

  20. Berezhkovskiy LM. Determination of volume of distribution at steady state with complete consideration of the kinetics of protein and tissue binding in linear pharmacokinetics. J Pharm Sci. 2004;93(2):364–74 (PubMed PMID: 14705193).

    Article  CAS  PubMed  Google Scholar 

  21. Piotrovski V, Van Peer A. A model with separate hepato-portal compartment (“first pass” model): fitting to plasma concentration-time profiles in humans. Pharm Res. 1997;14:230–7.

    Article  Google Scholar 

  22. Pang KS, Maeng HJ, Fan J. Interplay of transporters and enzymes in drug and metabolite processing. Mol Pharm. 2009;6:1734–55.

    Article  CAS  PubMed  Google Scholar 

  23. Maurer HH, Sauer C, Theobald DS. Toxicokinetics of drugs of abuse: current knowledge of the isoenzymes involved in the human metabolism of tetrahydrocannabinol, cocaine, heroin, morphine, and codeine. Ther Drug Monit. 2006;28(3):447–53.

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  25. Yang J, Jamei M, Yeo KR, Rostami-Hodjegan A, Tucker GT. Misuse of the well-stirred model of hepatic drug clearance. Drug Metab Dispos. 2007;35:501–2.

    Article  CAS  PubMed  Google Scholar 

  26. Park R, Leach WJ, Arieff AI. Determination of liver intracellular pH in vivo and its homeostasis in acute acidosis and alkalosis. Am J Physiol. 1979;236(3):F240–5.

    CAS  PubMed  Google Scholar 

  27. Thompson MD, Beard DA, Wu F. Use of partition coefficients in flow-limited physiologically-based pharmacokinetic modeling. J Pharmacokinet Pharmacodyn. 2012;39:313–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Heuberger JA, Guan Z, Oyetayo OO, et al. Population pharmacokinetic model of THC integrates oral, intravenous, and pulmonary dosing and characterizes short- and long-term pharmacokinetics. Clin Pharmacokinet. 2015;54(2):209–19.

    Article  CAS  PubMed  Google Scholar 

  29. Awasthi R, An G, Donovan MD, Boles Ponto LL. Relating observed psychoactive effects to the plasma concentrations of delta-9-tetrahydrocannabinol and its active metabolite: an effect-compartment modeling approach. J Pharm Sci. 2018;107:745–55.

    Article  CAS  PubMed  Google Scholar 

  30. Marsot A, Audebert C, Attolini L, Lacarelle B, Micallef J, Blin O. Population pharmacokinetics model of THC used by pulmonary route in occasional cannabis smokers. J Pharmacol Toxicol Methods. 2017;85:49–54.

    Article  CAS  PubMed  Google Scholar 

  31. Hendeles L, Weinberger M, Milavetz G, Hill M 3rd, Vaughan L. Food-induced, “dose-dumping” from a once-a day theophylline product as a cause of theophylline toxicity. Chest. 1985;87:758–65.

    Article  CAS  PubMed  Google Scholar 

  32. Lembeger L, Martz R, Rodda B, Forney R, Rowe H. Comparative pharmacology of delta-9-tetrahydrocannabinol and its metabolite, 11-OH-delta-9-tetrahydrocannabinol. J Clin Invest. 1973;52:2411–7.

    Article  Google Scholar 

  33. Hunt CA, Jones RT. Tolerance and disposition of tetrahydrocannabinol in man. J Pharmacol Exp Ther. 1980;215(1):35–44.

    CAS  PubMed  Google Scholar 

  34. Wall ME, Sadler BM, Brine D, Taylor H, Perez-Reyes M. Metabolism, disposition and kinetics of delta-9-tetrahydrocannabinol in men and women. Clin Pharmacol Ther. 1983;34:352–63.

    Article  CAS  PubMed  Google Scholar 

  35. Gillette JR, Pang KS. Theoretical aspects of pharmacokinetic drug interactions. Clin Pharmacol Ther. 1997;22:623–39.

    Article  Google Scholar 

  36. Lutz JD, Fujioka Y, Isoherranen N. Rationalization and prediction of in vivo metabolite exposures: the role of metabolite kinetics, clearance predictions and in vitro parameters. Expert Opin Drug Metab Toxicol. 2010;6(9):1095–109.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Yang F, Xiong X, Liu Y, et al. CYP2C9 and OATP1B1 genetic polymorphisms affect the metabolism and transport of glimepiride and gliclazide. Sci Rep. 2018;8(1):10994. https://doi.org/10.1038/s41598-018-29351-4 (Published 2018 Jul 20).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Brunet B, Doucet C, Venisse N, et al. Validation of large white pig as an animal model for the study of cannabinoids metabolism: application to the study of THC distribution in tissues. Forensic Sci Int. 2006;161:169–74.

    Article  CAS  PubMed  Google Scholar 

  39. Bonate PL. Nonlinear mixed effects models: practical issues. In: Bonate PL, editor. Pharmacokinetic–pharmacodynamic modeling and simulation. 2nd ed. New York: Springer; 2011. p. 303.

    Chapter  Google Scholar 

  40. Wilsey BL, Deutsch R, Samara E, et al. A preliminary evaluation of the relationship of cannabinoid blood concentrations with the analgesic response to vaporized cannabis. J Pain Res. 2016;9:587–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Cooper ZD, Haney M. Sex-dependent effects of cannabis-induced analgesia. Drug Alcohol Depend. 2016;167:112–20.

    Article  PubMed  PubMed Central  Google Scholar 

  42. LeFoll B, Tyndale RF. Cannabinoids: friend or foe? Clin Pharmacol Ther. 2015;97(6):528–31.

    Article  CAS  Google Scholar 

  43. D’Souza DC, Ranganathan M. Medical marijuana–is the cart before the horse? JAMA. 2015;313(24):2431–2.

    Article  PubMed  Google Scholar 

  44. Volkow ND, Baler RD, Compton WM, Weiss SRB. Adverse health effects of marijuana use. N Engl J Med. 2014;370(23):2219–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Hiratsuka M. Genetic polymorphisms and in vitro functional characterization of CYP2C8, CYP2C9, and CYP2C19 allelic variants. Biol Pharm Bull. 2016;39:1748–59.

    Article  CAS  PubMed  Google Scholar 

  46. Noble MJ, Hedberg K, Hendrickson RG. Acute cannabis toxicity. Clin Toxicol (Phila). 2019;24:1–8. https://doi.org/10.1080/15563650.2018.1548708.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work is dedicated to the memory of our colleague and friend, Hans Sachs (12.7.1948–10.3.2017), former head of the Forensic and Toxicologic Center, FTC GmbH, Munich, Germany. Doctor Sachs provided all analytic results of the THC data and contributed greatly to the manuscript. Without him, this manuscript would never have been written. Dr. Wolowich would also like to acknowledge personal conversations with KS Pang, WJ Jusko and Dr. Ryan Vandrey who graciously allowed the use of his oral cannabis study data. This work was presented, in part at the American College of Clinical Pharmacology, September, 2016 in Bethesda, MD, USA; the 8th Swiss Pharma Science Day, August, 2015 Bern, Switzerland; the Annual Meeting American Academy of Forensic Science, February, 2014, Seattle, WA, USA; the American Conference of Pharmacometrics Annual Meeting, May, 2013, Ft. Lauderdale FL, USA; and the American Society of Anesthesiology Annual Meeting, Chicago, IL, USA in October, 2011.

Author information

Authors and Affiliations

Authors

Contributions

WRW was responsible for writing the manuscript and all pharmacokinetic data analysis and mPBPK modeling. RG designed and conducted sampling of the probes, and revised the manuscript. MKB designed the study, helped with the conduct of the study and with writing of the manuscript. WB helped design the study, tested the quality of the THC solution used for the injection and coordinated the analysis of the probes. LT designed the study, and helped in writing the manuscript of this study.

Corresponding author

Correspondence to William R. Wolowich.

Ethics declarations

Funding

No source of funding was used for this study.

Conflict of interest

All authors have no conflict of interest to declare.

Ethics approval

All procedures performed in studies involving human participants were in accordance with the ethical standards of the Institutional Review Board approval (Cantonal Ethics Committee Bern Approval#: KEK 241–09, ISRCTN 53019164) and permission of all relevant bodies (BAG, Swissmedic) and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards.

Informed consent

Written informed consent was obtained from all volunteers.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary material 1 (PDF 695 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wolowich, W.R., Greif, R., Kleine-Brueggeney, M. et al. Minimal Physiologically Based Pharmacokinetic Model of Intravenously and Orally Administered Delta-9-Tetrahydrocannabinol in Healthy Volunteers. Eur J Drug Metab Pharmacokinet 44, 691–711 (2019). https://doi.org/10.1007/s13318-019-00559-7

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s13318-019-00559-7

Navigation