Conference PaperPDF Available

Assessment of the absorption of a standardized whole plant extract of cannabidiol and tetrahydrocannabinol in comparative ratios in patients with glioblastoma multiforme or high grade glioma.

Authors:

Abstract

Twenty participants (n=20) from from a phase II double blind randomised clinical control trial assessing the tolerability of medicinal cannabis in patients with stage 4 or recurrent glioblastoma multiforme (GBM) were assessed in a secondary data analysis. The primary clinical trial protocol has been designed in part to collect data for this projects' purpose. The aim was to assess two medicinal cannabis ratios [THC:CBD, 1:1 (5.8mg/ml:5.6mg/ml) and 4:1 (15mg/ml:3.8mg/ml)] and dosing to the measurable levels of plasma cannabinoids and cannabis metabolites collected at four time points via validated liquid chromatography-tandem mass spectrometry (LC-MS/MS) [5]. This assay was chosen to identify any correlation to medicinal cannabis ratio and the individual dose. Individual tolerability was subjectively measured via ADLs. Participants were instructed to titrate the dose of the intervention by 0.2ml/daily until an individual tolerability was reached. The analysis for absorption includes the results of each measured cannabinoid for every participants over the four time points using repeated measures. Difference in plasma cannabinoids levels over time was described using multiple linear regression to predict the value of the independent variable. Spearman's Correlation determined the strength and direction of a non-linear, monotonic relationship between variables. To examine the combined and the unique influences of the independent variables of BMI, weight, gender, age and dose a hierarchical regression over the 3 time points for each cannabinoid and cannabinoid metabolite was conducted. Medicinal cannabis was shown to have an individual dose tolerability, and that inter-individual variability from the dose input is influenced by multiple dynamic pharmacokinetic variables that also demonstrate intra-individual variance in cannabinoids and cannabinoid metabolites over time. There was a positive correlation between certain plasma cannabinoids and cannabinoid metabolites and the dose of medicinal cannabis at some time points, however the known influences of the pharmacokinetic parameters of metabolism in this population did not significantly predict plasma cannabinoid metabolites. There was a positive correlation between dose and plasma THC in linear regressions, however after controlling for age, gender, weight and BMI in hierarchical regression, the effect of dose was not significant at any time point. This finding suggests that dose is supported by the variables of age, gender, weight and BMI, but no one was more indicative than another. This is the first study of this type to examine the dose variability and metabolism of plasma cannabinoids and cannabinoid metabolites of an oral cannabis based medicine in patients with glioblastoma multiforme or high grade glioma. A secondary analysis of existing data from a Phase II double blind randomised clinical trial assessing the tolerability of medicinal cannabis in patients with high grade glioma or recurrent GBM (ACTRN 12617001287325) has been analysed to measure a correlation of orally administered medicinal cannabis ratios and dosing to the measurable levels of cannabinoids and cannabinoid metabolites. This project provides an unique insight on the pharmacokinetics of medicinal cannabis (MC) with respect to the absorption of two different ratios of MC and the individual dose effect variables in oncology.
Danielle Brown1, Michael Watson1, Janet Schloss1,2
1. Endeavour College of Natural Health, Australia 2. Australian Research Centre in Complementary and Integrative Medicine (ARCCIM), UTS, Australia
Twenty participants (n=20) from from a phase II double blind randomised clinical control trial
assessing the tolerability of medicinal cannabis in patients with stage 4 or recurrent
glioblastoma multiforme (GBM) were assessed in a secondary data analysis. The primary
clinical trial protocol has been designed in part to collect data for this projects’ purpose.
The aim was to assess two medicinal cannabis ratios [THC:CBD, 1:1 (5.8mg/ml:5.6mg/ml) and
4:1 (15mg/ml:3.8mg/ml)] and dosing to the measurable levels of plasma cannabinoids and
cannabis metabolites collected at four time points via validated liquid chromatography–tandem
mass spectrometry (LC-MS/MS)[5]. This assay was chosen to identify any correlation to
medicinal cannabis ratio and the individual dose. Individual tolerability was subjectively
measured via ADLs. Participants were instructed to titrate the dose of the intervention by
0.2ml/daily until an individual tolerability was reached.
The analysis for absorption includes the results of each measured cannabinoid for every
participants over the four time points using repeated measures. Difference in plasma
cannabinoids levels over time was described using multiple linear regression to predict the
value of the independent variable. Spearman’s Correlation determined the strength and
direction of a non-linear, monotonic relationship between variables. To examine the combined
and the unique influences of the independent variables of BMI, weight, gender, age and dose
a hierarchical regression over the 3 time points for each cannabinoid and cannabinoid
metabolite was conducted.
Medicinal cannabis was shown to have an individual dose tolerability, and that inter-individual
variability from the dose input is influenced by multiple dynamic pharmacokinetic variables
that also demonstrate intra-individual variance in cannabinoids and cannabinoid metabolites
over time.
There was a positive correlation between certain plasma cannabinoids and cannabinoid
metabolites and the dose of medicinal cannabis at some time points, however the known
influences of the pharmacokinetic parameters of metabolism in this population did not
significantly predict plasma cannabinoid metabolites.
There was a positive correlation between dose and plasma THC in linear regressions,
however after controlling for age, gender, weight and BMI in hierarchical regression, the effect
of dose was not significant at any time point. This finding suggests that dose is supported by
the variables of age, gender, weight and BMI, but no one was more indicative than another.
This is the first study of this type to examine the dose variability and metabolism of plasma
cannabinoids and cannabinoid metabolites of an oral cannabis based medicine in patients with
glioblastoma multiforme or high grade glioma. A secondary analysis of existing data from a Phase
II double blind randomised clinical trial assessing the tolerability of medicinal cannabis in patients
with high grade glioma or recurrent GBM (ACTRN 12617001287325) has been analysed to
measure a correlation of orally administered medicinal cannabis ratios and dosing to the
measurable levels of cannabinoids and cannabinoid metabolites.
This project provides an unique insight on the pharmacokinetics of medicinal cannabis (MC) with
respect to the absorption of two different ratios of MC and the individual dose effect variables in
oncology.
1. Russo, E.B. and J. Marcu, Cannabis Pharmacology: The Usual Suspects and a Few Promising Leads. Adv Pharmacol, 2017. 80: p. 67-134.
2. Tateo, S., State of the evidence: Cannabinoids and cancer pain-A systematic review. Journal of the American Association of Nurse Practitioners, 2017. 29(2): p. 94-103.
1. MacCallum, C.A. and E.B. Russo, Practical considerations in medical cannabis administration and dosing. European Journal of Internal Medicine, 2018. 49: p. 12-19.
4. Stout, S.M. and N.M. Cimino, Exogenous cannabinoids as substrates, inhibitors, and inducers of human drug metabolizing enzymes: a systematic review. Drug Metabolism Reviews, 2014. 46(1): p. 86-95.
5. Schwope, D. M., Scheidweiler, K. B., & Huestis, M. A. (2011). Direct Quantification of Cannabinoids and Cannabinoid Glucuronides in Whole Blood by Liquid Chromatography Tandem Mass Spectrometry. Analytical and bioanalytical chemistry,
401(4), 1273-1283. doi:10.1007/s00216-011-5197-7
6. Huestis, M.A., Human Cannabinoid Pharmacokinetics. Chemistry & biodiversity, 2007. 4(8): p. 1770-1804.
7. Sharma, P., P. Murthy, and M.M.S. Bharath, Chemistry, Metabolism, and Toxicology of Cannabis: Clinical Implications. Iranian Journal of Psychiatry, 2012. 7(4): p. 149-156.
8. Grotenhermen, F., Pharmacokinetics and pharmacodynamics of cannabinoids. Clinical Pharmacokinetics, 2003. 42(4): p. 327-360.
9. Turgeman, I. and G. Bar-Sela, Cannabis use in palliative oncology: A review of the evidence for popular indications. Israel Medical Association Journal, 2017. 19(2): p. 85-88.
10.. Piomelli, D. and E.B. Russo, The Cannabis sativa Versus Cannabis indica Debate: An Interview with Ethan Russo, MD. Cannabis Cannabinoid Res, 2016. 1(1): p. 44-46.
19 participants (n=19) with stage 4 GBM or recurrent high grade gliomas completed the 12
week trial. All participants reached individual tolerability by week 5. All but one participant who
was withdrawn continued on the individual tolerable dose until trial conclusion.
The average dose for intervention arm A was 11.14mg/ml THC and 11.54 CBD mg/ml.
Intervention arm B was 27.6mg/ml THC and 6.99mg/ml CBD. There was a positive correlation
between dose and plasma THC in linear regressions, however after controlling for age,
gender, weight and BMI in hierarchical regression, the effect of dose was not significant at
any time point. This finding suggests that dose is supported by the variables of age, gender,
weight and BMI, but no one was more indicative than another.
A statistically significant variance in plasma cannabinoid and cannabinoid metabolites can be
attributed to the different dosage for tetrahydrocannabinolic acid (THCA) (p=0.090),
THCCOOH (p=0.032) and THCCOOH-glucuronide (p=0.001) at week 4. With CBD, 74% and
89% of the variance in plasma cannabidiol was explained by dose at weeks 8 and 12
respectively. Participants reporting raised liver enzymes at baseline had normal range or
close to normal range liver enzymes by week 12.
0.0
5.0
10.0
15.0
20.0
25.0
1 2 3 4
ng/ml
Time Points (Initial, 4 Weeks, 8 Weeks, 12 Weeks)
Intervention A: THCA
MC002
MC004
MC007
MC012
MC013
MC015
MC019
MC022
MC025
MC029
0.0
5.0
10.0
15.0
20.0
25.0
30.0
1 2 3 4
ng/ml
Time Points (Initial, 4 Weeks, 8 Weeks, 12 Weeks)
Intervention B: THCA
MC001
MC006
MC011
MC012
MC016
MC018
MC020
MC023
MC024
MC027
0.0
0.5
1.0
1.5
2.0
2.5
3.0
3.5
1234
ng/ml
Time Points (Initial, 4 Weeks, 8 Weeks, 12 Weeks)
Intervention A: THC
MC002
MC004
MC007
MC012
MC013
MC015
MC019
MC022
MC025
MC029
0.0
1.0
2.0
3.0
4.0
5.0
6.0
7.0
1 2 3 4
ng/ml
Time Points (Initial, 4 Weeks, 8 Weeks, 12 Weeks)
Intervention B: THC
MC001
MC006
MC011
MC014
MC016
MC018
MC020
MC023
MC024
MC027
0.0
100.0
200.0
300.0
400.0
500.0
600.0
1234
ng/ml
Time Points (Initial, 4 Weeks, 8 Weeks, 12 weeks)
Intervention A: THCCOOH
MC002
MC004
MC007
MC012
MC013
MC015
MC019
MC022
MC025
MC029
0.0
100.0
200.0
300.0
400.0
500.0
600.0
1 2 3 4
ng/ml
Time Points (Initial, 4 Weeks, 8 Weeks, 12 weeks)
Intervention B: THCCOOH
MC001
MC006
MC011
MC014
MC016
MC018
MC020
MC023
MC024
MC027
0.0
500.0
1000.0
1500.0
2000.0
2500.0
3000.0
3500.0
1234
Time Points (Initial, 4 Weeks, 8 Weeks, 12 weeks)
Intervention A: THCCOOH-glucuronide
MC002
MC004
MC007
MC012
MC013
MC015
MC019
MC022
MC025
MC029
0.0
500.0
1000.0
1500.0
2000.0
2500.0
3000.0
3500.0
4000.0
4500.0
5000.0
1 2 3 4
ng/ml
Time Points (Initial, 4 Weeks, 8 Weeks, 12 weeks)
Intervention B: THCCOOH-glucoronide
MC001
MC006
MC011
MC014
MC016
MC018
MC020
MC023
MC024
MC027
0.0
10.0
20.0
30.0
40.0
50.0
60.0
70.0
80.0
1234
ng/ml
Time Points (Initial, 4 weeks, 8 Weeks, 12 Weeks)
Intervention A: THC-glucuronide
MC002
MC004
MC007
MC012
MC013
MC019
MC022
MC025
MC029
0.0
2.0
4.0
6.0
8.0
10.0
12.0
14.0
16.0
18.0
20.0
1 2 3 4
ng/ml
Time Points (Initial, 4 weeks, 8 Weeks, 12 Weeks)
Intervention B: THC-glucuronide
MC001
MC006
MC011
MC014
MC016
MC018
MC020
MC023
MC027
MC024
0.0
0.1
0.2
0.3
0.4
0.5
0.6
0.7
0.8
0.9
1 2 3 4
ng/ml
Time Points (Initial, 4 Weeks, 8 Weeks, 12 Weeks)
Intervention A: CBDA
MC002
MC004
MC007
MC012
MC013
MC015
MC019
MC022
MC025
MC029
0.0
0.1
0.2
0.3
0.4
0.5
0.6
0.7
0.8
0.9
1 2 3 4
ng/ml
Time Points (Initial, 4 Weeks, 8 Weeks, 12 Weeks)
Intervention B: CBDA
MC001
MC006
MC011
MC014
MC016
MC018
MC020
MC023
MC024
MC027
0.0
0.1
0.2
0.3
0.4
0.5
0.6
0.7
0.8
0.9
1234
ng/ml
Time Points (Initial, 4 Weeks, 8 Weeks, 12 Weeks)
Intervention A: CBD
MC002
MC004
MC007
MC012
MC013
MC015
MC019
MC022
MC025
MC029
0.0
0.1
0.2
0.3
0.4
0.5
0.6
0.7
0.8
0.9
1 2 3 4
ng/ml
Time Points (Initial, 4 Weeks, 8 Weeks, 12 Weeks)
Intervention B: CBD
MC001
MC006
MC011
MC014
MC016
MC018
MC020
MC023
MC027
MC024
Pharmacological constituents found in Cannabis sp have demonstrated both palliative and
modulatory effects in cancer management. An understanding of the administration, dose,
absorption, metabolism and potential drug interactions of cannabis-based medicines in cancer
populations is fundamental for the assessment of efficacy, safety and therapeutic index of
medicinal cannabis.
Pharmaceutical cannabis-based medicines and single molecule cannabinoids are approved for
use in several countries despite the common conclusion within the broader medical community
that medicinal cannabis lacks adequate evidence[3]. A dose titration approach is used to identify
an individual dose effect and to navigate the therapeutic window of a liquid medicinal cannabis to
decrease the risk of an adverse events and maintain a dose for efficacy. However, this results in a
high variability of individual dosing to achieve a therapeutic outcome.
Dose (mL)
Dose (mL)
Intervention A
Week 4
Week 8
Intervention B
Week 4
Week 8
Week 12
MC003
1
1
MC013
2.1
2.1
2.1
MC004
0.6
0.6
MC014
2
2
2
MC001
2.8
3
MC015
2.4
2.4
2.4
MC005
2.6
3
MC011
1.6
1.6
1.6
MC006
2.2
2.2
MC016
0.9
0.9
0.9
MC002
2
2
MC012
2.6
2.6
2.6
MC007
2.2
2.2
MC017
1.6
1.6
1.6
MC008
1.9
1.9
MC018
1.8
1.8
1.8
MC009
2
2
MC019
2
2
2
MC010
2
2
MC020
1.4
1.4
1.4
Individual plasma levels of cannabinoids and metabolites measured in nanograms per
millilitre (ng/ml) reported at baseline (initial) weeks 4, 8 and 12 for intervention A and B.
Reporting of dose titration (millilitres) of two ratios of medicinal cannabis (Group A, Group B)
reported at 3 times points, over 12-week trial enrolment
Results of this study identified an increased tolerability to a higher dose of THC when in
ratio with CBD. A whole plant cannabis extract may allow for higher dosing when when
establishing a therapeutic outcome associated with THC.
The association of raised liver enzymes and cannabis use is not well documented, however
in this cohort there is an absence of any rise in LFT markers across the 12-week period.
More research is required to measure if this observation is a direct or indirect association of
the intervention itself and/or reduced dexamethasone. Furthermore, it was identified that
participants who had raised liver enzymes at baseline had normal range or close to normal
range liver enzymes by week 12.
A continued understanding of the role of dose within therapeutic considerations of patient
centred outcomes in this population may support improved clinical management of patient
outcomes and quality of life.
There are multiple intersecting confounders to consider in cannabinoid metabolism
including the influence of concomitant medications and the direct influence they can have
on microsomal hepatic metabolism that can shift the parameters of individual tolerability,
and individual pharmacogenomic considerations that influence both cannabinoid
metabolism and phase I and phase II hepatic metabolism.
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Cannabis has been employed medicinally throughout history, but its recent legal prohibition, biochemical complexity and variability, quality control issues, previous dearth of appropriately powered randomised controlled trials, and lack of pertinent education have conspired to leave clinicians in the dark as to how to advise patients pursuing such treatment. With the advent of pharmaceutical cannabis-based medicines (Sativex/nabiximols and Epidiolex), and liberalisation of access in certain nations, this ignorance of cannabis pharmacology and therapeutics has become untenable. In this article, the authors endeavour to present concise data on cannabis pharmacology related to tetrahydrocannabinol (THC), cannabidiol (CBD) et al., methods of administration (smoking, vaporisation, oral), and dosing recommendations. Adverse events of cannabis medicine pertain primarily to THC, whose total daily dose-equivalent should generally be limited to 30mg/day or less, preferably in conjunction with CBD, to avoid psychoactive sequelae and development of tolerance. CBD, in contrast to THC, is less potent, and may require much higher doses for its adjunctive benefits on pain, inflammation, and attenuation of THC-associated anxiety and tachycardia. Dose initiation should commence at modest levels, and titration of any cannabis preparation should be undertaken slowly over a period of as much as two weeks. Suggestions are offered on cannabis-drug interactions, patient monitoring, and standards of care, while special cases for cannabis therapeutics are addressed: epilepsy, cancer palliation and primary treatment, chronic pain, use in the elderly, Parkinson disease, paediatrics, with concomitant opioids, and in relation to driving and hazardous activities.
Article
Full-text available
Background: A flowering plant of variegated ingredients and psychoactive qualities, cannabis has long been used for medicinal and recreational purposes. Currently, cannabis is approved in several countries for indications of symptomatic alleviation. However, limited knowledge on the benefits and risks precludes inclusion of cannabis in standard treatment guidelines. This review provides a summary of the available literature on the use of cannabis and cannabinoid-based medicines in palliative oncology. Favorable outcomes are demonstrated for chemotherapy-induced nausea and vomiting and cancer-related pain, with evidence of advantageous neurological interactions. Benefit in the treatment of anorexia, insomnia and anxiety is also suggested. Short- and long-term side effects appear to be manageable and to subside after discontinuation of the drug. Finally, cannabinoids have shown anti-neoplastic effects in preclinical studies in a wide range of cancer cells and some animal models. Further research is needed before cannabis can become a part of evidence-based oncology practice.
Article
Full-text available
Dr. Ethan Russo, MD, is a board-certified neurologist, psychopharmacology researcher, and Medical Director of PHYTECS, a biotechnology company researching and developing innovative approaches targeting the human endocannabinoid system. Previously, from 2003 to 2014, he served as Senior Medical Advisor and study physician to GW Pharmaceuticals for three Phase III clinical trials of Sativex® for alleviation of cancer pain unresponsive to optimized opioid treatment and studies of Epidiolex® for intractable epilepsy. He has held faculty appointments in Pharmaceutical Sciences at the University of Montana, in Medicine at the University of Washington, and as visiting Professor, Chinese Academy of Sciences. He is a past President of the International Cannabinoid Research Society and former Chairman of the International Association for Cannabinoid Medicines. He serves on the Scientific Advisory Board for the American Botanical Council. He is the author of numerous books, book chapters, and articles on Cannabis, ethnobotany, and herbal medicine. His research interests have included correlations of historical uses of Cannabis with modern pharmacological mechanisms, phytopharmaceutical treatment of migraine and chronic pain, and phytocannabinoid/terpenoid/serotonergic/vanilloid interactions.
Article
Full-text available
The first method for quantifying cannabinoids and cannabinoid glucuronides in whole blood by liquid chromatography–tandem mass spectrometry (LC–MS/MS) was developed and validated. Solid-phase extraction followed protein precipitation with acetonitrile. High-performance liquid chromatography separation was achieved in 16 min via gradient elution. Electrospray ionization was utilized for cannabinoid detection; both positive (Δ9-tetrahydrocannabinol [THC] and cannabinol [CBN]) and negative (11-hydroxy-THC [11-OH-THC], 11-nor-9-carboxy-THC [THCCOOH], cannabidiol [CBD], THC-glucuronide, and THCCOOH-glucuronide) polarity were employed with multiple reaction monitoring. Calibration by linear regression analysis utilized deuterium-labeled internal standards and a 1/x 2 weighting factor, yielding R 2 values >0.997 for all analytes. Linearity ranged from 0.5 to 50 μg/L (THC-glucuronide), 1.0–100 μg/L (THC, 11-OH-THC, THCCOOH, CBD, and CBN), and 5.0–250 μg/L (THCCOOH-glucuronide). Imprecision was <10.5% CV, recovery was >50.5%, and bias within ±13.1% of target for all analytes at three concentrations across the linear range. No carryover and endogenous or exogenous interferences were observed. This new analytical method should be useful for quantifying cannabinoids in whole blood and further investigating cannabinoid glucuronides as markers of recent cannabis intake. Figure LC–MS/MS MRM ion chromatograms of extracted blank whole blood and cannabinoid analytes at limits of quantification
Article
Full-text available
Delta(9)-Tetrahydrocannabinol (THC) is the main source of the pharmacological effects caused by the consumption of cannabis, both the marijuana-like action and the medicinal benefits of the plant. However, its acid metabolite THC-COOH, the non-psychotropic cannabidiol (CBD), several cannabinoid analogues and newly discovered modulators of the endogenous cannabinoid system are also promising candidates for clinical research and therapeutic uses. Cannabinoids exert many effects through activation of G-protein-coupled cannabinoid receptors in the brain and peripheral tissues. Additionally, there is evidence for non-receptor-dependent mechanisms. Natural cannabis products and single cannabinoids are usually inhaled or taken orally; the rectal route, sublingual administration, transdermal delivery, eye drops and aerosols have only been used in a few studies and are of little relevance in practice today. The pharmacokinetics of THC vary as a function of its route of administration. Pulmonary assimilation of inhaled THC causes a maximum plasma concentration within minutes, psychotropic effects start within seconds to a few minutes, reach a maximum after 15-30 minutes, and taper off within 2-3 hours. Following oral ingestion, psychotropic effects set in with a delay of 30-90 minutes, reach their maximum after 2-3 hours and last for about 4-12 hours, depending on dose and specific effect. At doses exceeding the psychotropic threshold, ingestion of cannabis usually causes enhanced well-being and relaxation with an intensification of ordinary sensory experiences. The most important acute adverse effects caused by overdosing are anxiety and panic attacks, and with regard to somatic effects increased heart rate and changes in blood pressure. Regular use of cannabis may lead to dependency and to a mild withdrawal syndrome. The existence and the intensity of possible long-term adverse effects on psyche and cognition, immune system, fertility and pregnancy remain controversial. They are reported to be low in humans and do not preclude legitimate therapeutic use of cannabis-based drugs. Properties of cannabis that might be of therapeutic use include analgesia, muscle relaxation, immunosuppression, sedation, improvement of mood, stimulation of appetite, antiemesis, lowering of intraocular pressure, bronchodilation, neuroprotection and induction of apoptosis in cancer cells.
Article
Background and purpose: Cannabinoids are widely used to alleviate intractable symptoms such as pain, nausea, and muscle spasticity. The purpose of this review was to ascertain the current state of the science regarding use of cannabinoids for cancer pain. Methods: Four electronic databases were searched for randomized control trials of cannabinoids and cancer pain. Studies included examined the analgesic effects of cannabinoids for cancer pain. Methodological quality was assessed using the Jadad scale. Conclusions: Eight randomized control trials met the inclusion criteria for review. Most trials found analgesic effects from cannabinoids when compared to placebo, although not all associations reached statistical significance. The analgesic effects of cannabinoids were also limited by dose-dependent side effects. Side effects most commonly reported were changes in cognition, sedation, and dizziness. Implications for practice: There is evidence that cannabinoids are effective adjuvants for cancer pain not completely relieved by opioid therapy, but there is a dearth of high-quality studies to support a stronger conclusion. Cannabinoids appear to be safe in low and medium doses. Methodological limitations of the trials limited the ability to make sound conclusions. Further research is warranted before efficacy, safety, and utility of cannabinoids for cancer pain can be determined.
Article
Abstract Exogenous cannabinoids are structurally and pharmacologically diverse compounds that are widely used. The purpose of this systematic review is to summarize the data characterizing the potential for these compounds to act as substrates, inhibitors, or inducers of human drug metabolizing enzymes, with the aim of clarifying the significance of these properties in clinical care and drug interactions. In vitro data were identified that characterize cytochrome P-450 (CYP-450) enzymes as potential significant contributors to the primary metabolism of several exogenous cannabinoids: tetrahydrocannabinol (THC; CYPs 2C9, 3A4); cannabidiol (CBD; CYPs 2C19, 3A4); cannabinol (CBN; CYPs 2C9, 3A4); JWH-018 (CYPs 1A2, 2C9); and AM2201 (CYPs 1A2, 2C9). CYP-450 enzymes may also contribute to the secondary metabolism of THC, and UDP-glucuronosyltransferases have been identified as capable of catalyzing both primary (CBD, CBN) and secondary (THC, JWH-018, JWH-073) cannabinoid metabolism. Clinical pharmacogenetic data further support CYP2C9 as a significant contributor to THC metabolism, and a pharmacokinetic interaction study using ketoconazole with oromucosal cannabis extract further supports CYP3A4 as a significant metabolic pathway for THC and CBD. However, the absence of interaction between CBD from oromucosal cannabis extract with omeprazole suggests a less significant role of CYP2C19 in CBD metabolism. Studies of THC, CBD, and CBN inhibition and induction of major human CYP-450 isoforms generally reflect a low risk of clinically significant drug interactions with most use, but specific human data are lacking. Smoked cannabis herb (marijuana) likely induces CYP1A2 mediated theophylline metabolism, although the role of cannabinoids specifically in eliciting this effect is questionable.
Article
ChemInform is a weekly Abstracting Service, delivering concise information at a glance that was extracted from about 200 leading journals. To access a ChemInform Abstract, please click on HTML or PDF.
Cannabis Pharmacology: The Usual Suspects and a Few Promising Leads
  • E B Russo
  • J Marcu
Russo, E.B. and J. Marcu, Cannabis Pharmacology: The Usual Suspects and a Few Promising Leads. Adv Pharmacol, 2017. 80: p. 67-134.
  • P Sharma
  • P Murthy
  • M M S Bharath
  • Chemistry
  • Metabolism
  • Toxicology
  • Cannabis
Sharma, P., P. Murthy, and M.M.S. Bharath, Chemistry, Metabolism, and Toxicology of Cannabis: Clinical Implications. Iranian Journal of Psychiatry, 2012. 7(4): p. 149-156.