Next Article in Journal
Vanillin Promotes Osteoblast Differentiation, Mineral Apposition, and Antioxidant Effects in Pre-Osteoblasts
Next Article in Special Issue
In Vitro Stability and Pharmacokinetic Study of Pedunculoside and Its Beta-CD Polymer Inclusion Complex
Previous Article in Journal
Engineering Nanomedicine for Non-Viral RNA-Based Gene Therapy of Glioblastoma
Previous Article in Special Issue
Pharmacokinetic Interaction of Kratom and Cannabidiol in Male Rats
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

CBD and THC in Special Populations: Pharmacokinetics and Drug–Drug Interactions

by
Lixuan Qian
1,
Jessica L. Beers
2,†,
Klarissa D. Jackson
2 and
Zhu Zhou
1,*
1
Department of Chemistry, York College, City University of New York, Jamaica, NY 11451, USA
2
Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
*
Author to whom correspondence should be addressed.
Current address: Department of Pharmaceutics, University of Washington School of Pharmacy, Seattle, WA 98195, USA.
Pharmaceutics 2024, 16(4), 484; https://doi.org/10.3390/pharmaceutics16040484
Submission received: 18 February 2024 / Revised: 13 March 2024 / Accepted: 25 March 2024 / Published: 1 April 2024
(This article belongs to the Special Issue Pharmacokinetics, Pharmacodynamics and Drug Interactions)

Abstract

:
Cannabinoid use has surged in the past decade, with a growing interest in expanding cannabidiol (CBD) and delta-9-tetrahydrocannabinol (THC) applications into special populations. Consequently, the increased use of CBD and THC raises the risk of drug–drug interactions (DDIs). Nevertheless, DDIs for cannabinoids, especially in special populations, remain inadequately investigated. While some clinical trials have explored DDIs between therapeutic drugs like antiepileptic drugs and CBD/THC, more potential interactions remain to be examined. This review summarizes the published studies on CBD and THC–drug interactions, outlines the mechanisms involved, discusses the physiological considerations in pharmacokinetics (PK) and DDI studies in special populations (including pregnant and lactating women, pediatrics, older adults, patients with hepatic or renal impairments, and others), and presents modeling approaches that can describe the DDIs associated with CBD and THC in special populations. The PK of CBD and THC in special populations remain poorly characterized, with limited studies investigating DDIs involving CBD/THC in these populations. Therefore, it is critical to evaluate potential DDIs between CBD/THC and medications that are commonly used in special populations. Modeling approaches can aid in understanding these interactions.

1. Introduction

1.1. Brief Overview of CBD and THC

“Cannabis” refers to several plants of the genus Cannabis (family Cannabaceae), primarily C. sativa, which contains > 500 constituents, including > 100 cannabinoids [1,2,3]. The two most extensively studied cannabinoids are the non-psychotoxic cannabidiol (CBD) and the psychoactive delta-9-tetrahydrocannabinol (THC) [4]. The psychoactive effects of cannabinoids are mainly mediated by cannabinoid receptor 1 (CB1) receptors [5], and THC is the agonist of both CB1 and cannabinoid receptor 2 (CB2) [6]. CBD is considered non-psychotoxic, as it does not bind to the cerebral CB1 receptor [7,8]. Instead, it exerts a negative allosteric modulation on CB1 [8,9]. CBD is a low-affinity agonist of the CB2 receptor [10]. Since the first approval of CBD and the synthetic version of THC by the FDA, there has been a growing interest in CBD and THC clinical trials to treat epilepsy, pain, depression, anxiety, psychosis, neurodegenerative disease, gastrointestinal diseases, and addiction [10,11]. CBD was the top-selling herbal supplement ingredient in the US natural channel from 2018 to 2020, increasing by >450% from 2017 [12]. Purified CBD (brand name: Epidiolex®) has been approved in the United States and European Union for the treatment of refractory epileptic seizures in Dravet and Lennox–Gastaut patients, as well as seizures associated with tuberous sclerosis complex [13,14,15]. Meanwhile, cellular and rodent studies also suggest that CBD has therapeutic potential as a cardioprotective and anti-inflammatory agent [16,17,18]. The mean THC content in cannabis-derived products was reported to increase 2-fold from 2009 to 2017 [19]. To date, the U.S. Food and Drug Administration (FDA) has approved two drugs containing a synthetic version of THC (dronabinol) and one drug that is a synthetic analog of THC (nabilone). Both are indicated for nausea and vomiting associated with cancer chemotherapy; dronabinol is also indicated for anorexia associated with weight loss in patients with acquired immunodeficiency syndrome (AIDS) [20,21]. The 1:1 mixture of purified THC and CBD as an oromucosal spray (brand name: Sativex®) has been approved in the United Kingdom and other countries for the treatment of multiple sclerosis [22,23]. CBD/THC on the market and in development have various routes of administration, including oral, inhaled/vaporized, sublingual, and as an oromucosal spray [10].

1.2. Importance of Understanding Pharmacokinetics and Drug Interactions

Both therapeutic effects and adverse effects increase with exposure to CBD [24,25]. The proportion of patients experiencing transaminase elevations that are greater than three times the upper limit of the reference range in phase III trials increased from 6.8% at a CBD dose of 10 mg/kg/day to 27.7% at a CBD dose of 20 mg/kg/day, compared to 0.0% for the placebo [25]. THC doses above 10 mg are reported to be associated with an increased risk of addiction and THC-related psychosis [26,27]. For example, in a study in the UK, higher-potency cannabis users reported being four times more likely to have cannabis use problems than lower-potency cannabis users [28]. Pichini et al. suggested that smoking four cannabis cigarettes is associated with more severe drowsiness than smoking one cigarette [29]. Therefore, it is crucial to understand the pharmacokinetics (PK) of CBD and THC to determine the appropriate dose, maximize the treatment effect, and minimize the risk of adverse effects and addiction. However, the PK of CBD and THC exhibits great inter-individual variability in absorption, distribution, and metabolic clearance [4,30,31].
CBD is classified as a class II Biopharmaceutical Classification System (BCS) substance [32]. Its low water solubility and high passive permeability lead to poor and erratic oral absorption. CBD’s estimated absolute oral bioavailability is 6% with an empty stomach [33], increasing to 14–23% with food [34]. CBD’s relatively low oral bioavailability can be attributed to incomplete gastrointestinal absorption and extensive first-pass metabolism [33]. Distribution and elimination also play crucial roles in CBD’s highly variable PK. CBD’s high lipophilicity leads to a large volume of distribution and substantial accumulation in the adipose tissue. The volume of distribution (Vd) of CBD in adults ranged from 2.5 to 10 L/kg [35]. CBD concentrations are sensitive to body weight and adiposity. CBD’s metabolism involves both cytochromes P450 (CYP) and UDP-glucuronosyltransferases (UGT) [36,37,38]. Polymorphisms in these metabolic enzymes may introduce variability in PK, which will be discussed in Section 2.3. In plasma, the terminal elimination half-life (t1/2) of CBD varied from 24 to 61 h [35]. Most CBD and its metabolites are excreted through feces, and a limited part is excreted through urine [4].
Similar to CBD, THC is classified as a BCS class II drug [32], exhibiting limited and variable oral absorption, with an oral bioavailability of 4 to 20% [39]. THC is also widely distributed in fat and has a large Vd, about 10 L/kg [39]. Unlike CBD, THC undergoes metabolism solely through CYP enzymes, including CYP2C9, 2C19, and 3A4, although THC metabolites are glucuronidated [38,40,41]. Changes in the activity of these CYP enzymes could impact the PK of THC. Furthermore, sex differences in the PK of THC have been observed in both animal and clinical trials [42,43]. THC has a long terminal t1/2 of about 22 h and is excreted in feces and urine as glucuronidated metabolites [4].
As CYP enzymes contribute to the metabolism of both CBD and THC, medications affecting these enzymes could potentially lead to drug–drug interactions (DDIs). Therefore, a comprehensive understanding of drug interactions is crucial for optimizing therapeutic outcomes. This knowledge guides dosage adjustments and helps mitigate potential risks associated with variable drug responses.

1.3. Significance of Studying Special Populations

Another challenge in determining CBD and/or THC doses lies in the fact that the current PK knowledge of CBD and THC is derived from studies conducted on healthy adults. The impact of different factors, including but not limited to aging, obesity, and disease conditions, has not been fully assessed. For example, due to the highly lipophilic nature of CBD/THC, the distribution may be different among the pediatric population, older adults, and individuals with obesity. While prior publications have reviewed the PK of CBD and THC in healthy adults [4,44,45,46,47], there is a significant gap in current understanding when it comes to special populations. This review aims to address this gap by focusing on the PK and potential DDIs of CBD and THC in special populations.

2. CBD/THC PK-Based Drug Interactions

2.1. Overview of Drug Interactions Involving CBD and THC

With the increasing use of recreational and medical cannabis, drug interactions involving CBD and THC have raised concerns. While Sativex® was approved in the UK in 2011, widespread attention to the issue of DDIs involving CBD and/or THC began with the approval of Epidiolex®, which is used in the treatment for certain seizures in children and adults. It is, therefore, not surprising that most DDI studies have focused on investigating the interaction between CBD/THC and antiepileptic drugs in pediatric, healthy adults, or young seizure patients [48,49,50,51,52]. In pediatrics, the DDI between CBD and antiepileptic drugs, including stiripentol [50], valproate [50,51], and clobazam [51,52,53], was tested and validated. In young adults, antiepileptic drugs (including stiripentol [48,50], valproate [48,50,51], tiagabine [54], and clobazam [48,49,51]), as well as warfarin [55,56,57,58,59], clozapine [60], buprenorphine [61], tacrolimus [62], sirolimus [63], everolimus [63,64], caffeine [65], citalopram [66], and theophylline [67,68], have been reported as victims of CBD/THC according to clinical trials and/or case reports. Conflicting results were observed in the DDI effect of CBD and THC on phenobarbital [51,69]. Also, the effect of typical DDI perpetrators, including ketoconazole and rifampicin, on CBD and THC was assessed in young adults [48,70].
In addition to healthy adults, some studies have reported potential DDIs between CBD/THC and certain medications in special populations. For instance, interactions were noted with medications like clopidogrel that was co-administered with aspirin (76-year-old male) in older adults [71]. A recently published systematic review concluded that a DDI between warfarin and CBD/THC is probable, although the available evidence was limited to only seven published case reports [72]. In a separate trial involving human immunodeficiency virus (HIV)-infected patients, no interaction was reported between THC and indinavir or nelfinavir [73]. Additionally, investigations into potential DDIs between CBD/THC and addictive substances, such as methadone [74], fentanyl [75], morphine [76], and cocaine, were conducted [77]. Vierke et al. also characterized the DDI between buprenorphine and CBD/THC [61]. Additional studies on drug interactions involving CBD and THC in special populations will be discussed in the subsequent sections.

2.2. Mechanisms of CBD and THC Interactions with Other Drugs

The mechanisms of DDIs involving CBD/THC are primarily PK-based. Major phase I metabolizing enzymes play key roles, particularly CYP3A4, CYP2C9, and CYP2C19. As a result, DDIs can occur when an inhibitor and/or a substrate of these CYP enzymes is co-administered with CBD/THC. The major metabolizing enzymes and metabolites of CBD are summarized in Table 1. CBD is metabolized mainly to 7-OH-CBD by CYP2C9 and CYP2C19, while CYP3A4 is crucial for other hydroxylation products [37,78]. 7-OH-CBD is further metabolized to the inactive 7-COOH-CBD. The primary metabolizing enzyme for this step remains unidentified, although recent evidence suggests that both CYP and aldehyde dehydrogenase (ALDH) enzymes are involved [79]. 7-OH-CBD and 7-COOH-CBD can undergo glucuronidation in phase II metabolism, but the specific UGTs that are involved require further study. Inducers and inhibitors of the metabolic enzymes (as listed in Table 1) have the potential to interact with CBD and its metabolites. Studies present inconsistent conclusions regarding whether CBD is a substrate for transporters. In silico studies suggest that CBD acts as a P-glycoprotein (P-gp, also known as multidrug resistance protein 1, MDR1, or ATP-binding cassette subfamily B member 1, ABCB1) substrate [80], while animal studies propose that CBD is not a substrate for P-gp or the breast cancer resistance protein (BCRP) [81]. In vitro, the major metabolite 7-COOH-CBD is a substrate for P-gp and an inhibitor of BCRP and the bile salt export pump [14]. A study from Holland et al. found that CBD strongly inhibited ATP-binding cassette subfamily C member 1 (ABCC1, also known as multiple-drug-resistant transporters 1, MRP1) in ovarian carcinoma cells overexpressing MRP1 [82]. However, to the authors’ knowledge, this has not been investigated in other cell types.
CBD carries the potential for numerous drug-metabolizing enzyme-mediated DDIs, and multiple instances of these interactions have been reported in the literature [51,85,87]. Enzymes that are impacted by CBD and the related parameters from in vitro studies are listed in Table 1. Current Epidiolex® prescribing information advises dosage adjustment of CBD with strong inducers of CYP3A4 or CYP2C19, as well as monitoring patients receiving concomitant substrates of CYP1A2, CYP2C8, CYP2C19, UGT1A9, or UGT2B7 for adverse effects associated with an inhibited metabolism of these substrates [14]. For transporters, Alsherbiny et al. noted in their review that the P-gp expression is sensitive to the duration of CBD exposure, being down-regulated in chronic exposure and up-regulated in short-term exposure [88].
THC is the substrate of CYPs in the phase I metabolism [41,83,84,89]. The major metabolizing enzymes and metabolites of THC are summarized in Table 1. A portion of the active main metabolite, 11-OH-THC, undergoes glucuronidation by UGT1A1 and UGT1A3 in the phase II metabolism [41,79,83,84]. The metabolite 11-COOH-THC is a substrate of the UGT1A family, primarily UGT1A9 and UGT1A10, in phase II metabolism [38]. Similar to CBD, there are conflicting results regarding whether THC is a substrate of transporters. Animal studies suggested that THC is the substrate of P-gp and BCRP [90,91]. In contrast, Chen et al. concluded from a Madin–Darby Canine Kidney cell study and an animal study that THC and most of its metabolites are not substrates of P-gp and BCRP, except for 11-COOH-THC, which is a poor substrate of BCRP [92,93].
Despite being the substrate of enzymes such as CYPs and UGTs, THC and its metabolites are also inhibitors of CYPs [65,85], MRP1 [82], and carboxylesterase 1 (CES1) in vitro [94]. According to another in vitro study, 11-COOH-THC is an inhibitor of BCRP [92]. Substrates of such enzymes are potential victims in DDIs involving THC. Enzymes that are impacted by THC and the related parameters from in vitro studies are listed in Table 1. Brzozowska et al. proposed that THC might increase the P-gp expression at the blood–brain barrier, leading to a decrease in the concentration of risperidone in the brain [95].
Many clinical trials have demonstrated DDIs with CBD and THC as perpetrators. Vierke et al. found that the CYP3A4 substrates buprenorphine and norbuprenorphine’s concentration were 2.7-fold and 1.4-fold higher in cannabis users [61]. When receiving a daily CBD dose of 2000–2900 mg, a 3-fold increase in normalized tacrolimus (CYP3A substrate) concentrations was reported by Leino et al. [62]. According to Jusko et al., when volunteers were exposed to cannabis, the clearance of theophylline (a CYP1A2 substrate) increased from 51.8 mL/h/kg to 73.3 mL/h/kg [68]. Thai et al. suggested that when another CYP1A2 substrate, caffeine, was co-administered with CBD (750 mg twice a day (b.i.d.)), the area under the curve (AUC) of caffeine increased by 95% [65]. One of the most studied CBD-induced DDIs is with clobazam, an antiepileptic drug that is often co-prescribed with CBD in patients with Lennox–Gastaut syndrome [48]. Clobazam is metabolized to the active metabolite N-desmethylclobazam by CYP3A4, and further hydroxylation to the inactive metabolite 4′-hydroxydesmethylclobazam is primarily mediated by CYP2C19 [96]. The inhibition of these P450 enzymes by CBD has been shown to significantly increase the exposure to N-desmethylclobazam, which has been demonstrated to increase the risk of adverse effects, including sedation, ataxia, irritability, tremor, urinary retention, and loss of appetite [48,51,53]. In a clinical trial involving ten male and ten female patients with epilepsy, CBD increased the AUC of the dosing interval and the Cmax of N-desmethylclobazam by 2.6-fold and 2.2-fold, respectively, when administered at a dose of 20 mg/kg/day [49]. As mentioned previously, there are also multiple case reports suggesting that cannabis can decrease the anticoagulant effect of warfarin, a CYP2C9 substrate [56,57,58,59,97,98]. Of note, CBD can act as a perpetrator of DDIs when combined with THC by inhibiting the metabolism of THC and 11-OH-THC. A recent clinical DDI study with eleven males and seven females found that orally administered CBD increased the oral exposure to THC and its metabolites 11-OH-THC and 11-COOH-THC when co-administered with a CYP probe drug cocktail [99]. Furthermore, patients receiving both CBD and THC experienced increased self-reported anxiety, sedation, and memory difficulties, as well as increased impairment of psychomotor and cognitive function compared to THC alone [99]. These clinical trials suggest that when co-administered with CYP2C9, 2C19, and 3A4 substrates, CBD and THC can lead to clinically significant DDIs (change in AUC ≥ 25% for inhibition, according to FDA guidance [100]) when acting as perpetrators. However, some clinical trials showed that THC did not inhibit the metabolism of some CYP2C19, 2C9, and 3A4 substrates as expected. Substrates of the enzymes that THC can inhibit, including the CYP3A4 and CYP2C19 substrate nelfinavir [73]; the CYP3A4 substrates indinavir [73], irinotecan [101], docetaxel [101], and doxorubicin [102]; and cyclophosphamide [102], a substrate of CYP2C9, 2C19, and 3A4, have been reported not to interact with THC. For example, the metabolism of nelfinavir (a substrate of CYP2C19 and 3A4) and indinavir (a substrate of CYP3A4) was expected to be inhibited by THC, as THC was reported as a CYP2C19 and 3A4 inhibitor in vitro, but a clinical trial reporting the statistically non-significant steady-state AUC decreases of 10.2% (p = 0.15) and 14.5% (p = 0.074), respectively, disproved this hypothesis [73]. In a clinical trial involving 14 male and 10 female cancer patients, there was no significant increase in the AUC with cannabis administered as an herbal tea for irinotecan (1.04-fold) or docetaxel (1.11-fold), even though irinotecan and docetaxel are mainly metabolized via CYP3A [101]. Similarly, Riggs et al. suggested that when used with THC, no DDI effect on the exposure to doxorubicin (a CYP3A4 substrate) and cyclophosphamide (a CYP2C9, 2C19, and 3A4 substrate) was observed. CBD is suggested to inhibit CES1 in vitro, but methylphenidate, a substrate of CES1, was reported to have no interaction with CBD [103]. More studies are necessary to investigate the mechanisms involved in these interactions between CBD/THC and drugs.
In addition to enzyme-mediated DDIs, several other factors might impact the PK of CBD and THC, as listed in Table 1. Due to its high lipophilicity, THC can accumulate in adipose tissue. Drugs that alter the total fat mass, fat distribution, or fat composition, such as glucocorticoids and diet pills, may influence the distribution of THC. Furthermore, 95–99% of plasma THC is bound to plasma proteins. Hence, competitive protein binding with other drugs may also change the PK of THC [104,105]. Additionally, there is controversy over whether CBD can be converted to THC in the body [106]. Some in vitro and animal studies suggested that CBD can be transformed into THC in simulated gastric fluid and rats [107,108]. However, a clinical trial designed specifically for the bioconversion of CBD to THC did not find such conversion [109].

2.3. Genetic Variations May Impact CBD/THC PK and Drug Interactions

Some DDIs that have been identified in vitro studies are deemed not clinically significant when validated in vivo. The differences between an in vitro prediction and the in vivo results may be explained by the drug–drug–gene interaction (DDGI) [110]. A DDGI can boost the DDI by affecting the metabolism pathways. However, a DDGI can also lead to a contrary effect with the DDI and diminish the DDI. The metabolism phenotype of the enzymes can lead to different types of DDGIs when there is an inhibitor or inducer [110]. Bahar et al. summarized the DDGIs on CYP2C9, CYP2C19, and CYP2D6. They suggested that the normal metabolizer phenotype leads to the greatest distortion of the DDIs, followed by the intermediate metabolizer and the least in the poor metabolizer [111]. CYP2C9, CYP2C19, and CYP2D6 are involved in the metabolism of 12.8%, 6.8%, and 20% of the majority of clinical drugs, respectively [112]. Their polymorphisms are the most frequently occurring variants in the phase I metabolism of drugs [113]. Bland et al. investigated the effects of polymorphism on CYP2C9 and found that the clearance for recombinantly expressed CYP2C9*2 and CYP2C9*3 were only 30% of that of the CYP2C9*1 (wild-type) [89]. Wolowich et al. investigated the effect of CYP2C9 polymorphism using physiologically based pharmacokinetic (PBPK) modeling and identified significant differences (p = 0.02) in the THC clearance between CYP2C9*3 homozygotes and other phenotypes. The total clearance of patients with CYP2C9 (homozygous/wild-type) was 41.9% of CYP2C9 (heterozygous/heterozygous) [114]. Additionally, Wolowich et al. also found that subjects who are homozygous for CYP2C9*3 have a 74% decrease in their THC-COOH exposure [114]. When predicting DDIs involving CYP2C9 using only wild-type CYP2C9, the contribution of the enzyme may be overestimated, leading to biased results. Given that CBD and THC can be metabolized by multiple CYPs, the importance of the second pathway will increase when the primary enzyme is reduced by a polymorphism. A more significant change in PK may be observed when both the primary and second pathways are influenced by DDIs, which may occur in polypharmacy.

3. CBD/THC Studies in Special Populations

In this section, studies on the effects of CBD and THC in special populations, including pregnant and lactating women, pediatrics, older adults, patients with hepatic or renal impairment, cancer patients, patients with neurological disorders, and others, are reviewed. We searched three databases (The Drug Interaction Database (DIDB®), PubMed, and Google Scholar) from inception until March 2024 to identify clinical trials on CBD/THC in special populations. The search terms included “CBD or THC” combined with the specific populations of interest. Trials that did not provide detailed PK parameters of CBD or THC in special populations were excluded from the tables but were still described in subsequent sections. Table 2 and Table 3 summarize the PK parameters of CBD and THC in clinical trials for special populations, respectively. PK parameters in healthy volunteers were not included as a reference due to variations in administration routes and a high PK variability.

3.1. Pregnancy and Lactation

3.1.1. Overview of Research on CBD/THC Use during Pregnancy and Lactation

The prevalence of self-reported past-month cannabis use among pregnant women in the US rose from 3.4% in 2003 to 7.0% in 2017 [127]. A study in California also showed that 19% of pregnant women aged 18 to 24 years used cannabis in 2016 [128]. Despite these trends, a limited number of studies have investigated the PK of CBD and THC and their metabolites in pregnant or lactating women. And there is a lack of cannabinoid plasma concentration–time data for pregnant or lactating women [129]. The FDA mandates postmarketing pregnancy outcomes research on Epidiolex®, and future research may rely on any forthcoming data from pregnant patients taking CBD who are enrolled in registries [130]. Metz et al. tested the serum, urine, and umbilical cord concentration of 11-COOH-THC in pregnant women from the gestational age of 16 weeks to delivery. Although the serum concentrations of 11-COOH-THC were measured, the trial failed to determine the THC concentrations [131]. Westin et al. reported that 11-COOH-THC was still detectable at week 14 after the last THC administration in one pregnant woman [132]. Baker et al. detected the THC levels in the breast milk of eight lactating women, suggesting that a mean of 2.5% of the THC dose was transferred into breast milk [133]. A 2018 study noted observable differences among THC, 11-OH-THC, and CBD in the breast milk of lactating women who used cannabis. THC was detected in the breast milk of 63% of the mothers, whereas only 9.0% showed detectable levels for either 11-OH-THC or CBD [134]. This pattern was also observed in a recent study, where 13 out of 14 breast milk samples showed THC levels, while detectable levels of 11-OH-THC and CBD were found in 3 and 1 sample, respectively. Additionally, this research reported that cannabis use also alters the lactose- and soluble IgA levels in the milk. Other CBD and THC studies in pregnant women and their infants focus on qualitative research on the adverse effects of CBD/THC on infants, e.g., malformations and effects on the development of male gonads [135,136]. No DDI studies involving CBD or THC in pregnant or lactation women were found. A PBPK model of THC was established by Patilea-Vrana et al. and extrapolated to pregnant women. The simulations showed no clinically significant change in the AUC0–t or Cmax of THC between pregnancy and nonpregnancy, but the AUC0–t and Cmax of 11-OH-THC decreased by up to 41% and 44%, respectively, during pregnancy compared with nonpregnancy [137]. However, this extrapolation was not validated due to a lack of clinical data. Shenkoya et al. developed and verified a paired lactation and infant THC PBPK model. When the authors simulated the inhalation of 0.045 g of THC, they found that the mean AUC and Cmax of breastmilk were 2.2 and 3.4 times higher than that of plasma, respectively [138].

3.1.2. PK Considerations in Pregnant and Lactating Women

Pregnant and lactating women undergo physiological changes that may result in altered PK of CBD or THC. However, the overall impact of pregnancy and lactation on CBD and THC exposure remains unknown. An individual’s body fat increases during pregnancy due to increased adipogenesis in the adipose tissue and adipocyte hyperplasia [139]. The fat stores in pregnant women reach their peak towards the end of the second trimester and then diminish [139,140]. Consequently, there may be changes in the volume of distribution of CBD/THC and their metabolites during pregnancy, potentially leading to variations in concentrations with gestation. Cardiovascular changes occur during pregnancy, including an increase in heart rate, a rise in plasma volume, an increase in plasma lipids, and a decrease in plasma protein levels. The increase in plasma volume may result in lower plasma CBD/THC concentrations, while the reduction in plasma protein levels may exacerbate the concentration decrease. In contrast, the free cannabinoid concentrations may increase due to the decreased plasma protein level, as CBD and THC have high protein binding (95–99% for THC) [104,105]. For elimination, Grant et al. suggested that the clearance of THC during pregnancy can increase up to 2-fold, and the THC exposure may be decreased [141]. Lastly, the cannabinoid concentration in non-blood matrices can be a good alternative. However, data interpretation could be challenging due to a lack of information [142].

3.1.3. Potential Drug Interactions and Safety Concerns for this Population

The use of cannabis by pregnant women should be carefully considered, as prenatal cannabis use can be associated with potential harm to the infant, including a lower birth weight, preterm birth, neonatal intensive care unit admission, and attention and externalizing problems [143]. However, due to the high utilization rate and the unavoidable passive cannabis intake, future studies may focus on how to decrease the harm of exposure to cannabis during pregnancy. Additionally, physiologic changes during pregnancy, such as hypertension during pregnancy and gestational diabetes, may necessitate the use of medications for some pregnant women. There is a knowledge gap in DDI studies between related drugs and CBD/THC, and further study is required.

3.2. Pediatrics

3.2.1. Overview of Studies Investigating CBD/THC in Pediatric Populations

Since the FDA approval of purified cannabidiol in the treatment of pediatric epilepsy, there has been a surge in research on the medical uses of CBD in pediatrics [144]. Most studies focus on the pharmacodynamics (PD) and the potential treatment effect of CBD and THC. In addition to the approved use for treatment-resistant epilepsy in pediatrics, research has explored the effect of CBD in oncology [145], autism spectrum disorder [146], severe behavioral problems in children with intellectual disability [147], fragile X syndrome [148], and complex motor disorder [149]. One clinical trial found that THC is a significant anti-nausea and anti-vomiting agent in the pediatric population. In some cases, THC has been shown to enhance individuals’ appetite [150]. The positive THC treatment effects were further investigated in complex motor disorders and spasticity [149,151]. However, adverse effects such as fatigue, sedation, nausea, diarrhea, appetite suppression, somnolence, and hallucinations were reported in the clinical CBD trials [152,153]. Additionally, Smid et al. suggested that THC-COOH exposure was associated with worse attention scores in early childhood [154].
PK descriptions of CBD and THC in pediatrics are limited. Wheless et al. characterized the high inter-individual variability in systemic cannabidiol exposure in pediatric patients with treatment-resistant epilepsy, and the PK parameters can be found in Table 2. They further noted that the inter-individual variability decreased during the long-term use of CBD and that CBD exposure in infants was approximately 40% of that in children or adolescents [52]. Other studies also reported a large inter-individual variability of over 80% in all PK parameters except for Tmax in CBD exposure. They observed a disproportionate increase in CBD exposure with ascending dose over a clinically relevant dose range from 5 to 20 mg/kg/day (Table 2) [115,155]. In contrast, Devinsky et al. and Wheless et al. suggested a linear relationship between exposure doses within the same dose range [52,156]. Wang et al. found that the time to peak plasma CBD concentrations and the mean acute elimination half-life were both shorter in the pediatric population than in adults [116]. Crockett et al. investigated the intra-individual variability of THC and suggested that it ranged from moderate to high (coefficient of variation [CV] 20–121%) [157]. No PK studies of THC in pediatrics have been reported.
Bansal et al. developed and verified a PBPK model to predict systemic CBD exposure in children aged 4–10 years, suggesting that pediatric patients have a comparable simulated CBD accumulation ratio to adults. In their models, the estimated fraction of CBD escaping the liver and plasma clearance in pediatrics was 155% and 63% of adults, respectively, and involving the ontogeny of UGT2B7 for children in the model made a minimal effect on the AUC and Cmax of CBD [158]. Shenkoya et al. also simulated the dose of THC in breast milk and infant THC exposure with their paired lactation and infant THC PBPK model. They predicted that 0.34% to 0.88% of the mother’s THC dose was delivered to the infant, and the mother–infant plasma THC AUC0–24h ratio increased to three times (3.4–3.6) with a six-fold increase in the amount of THC that was smoked by the mother [138].

3.2.2. PK Profiles and Considerations in Pediatric Patients

The PK of pediatric patients could be complex due to the child’s growth and development process. No significant differences in CBD exposure between children and adolescents were observed in a clinical trial, while the CBD exposure in infants was also lower than in adolescents [52].
When administered orally, the first-pass metabolism of a drug is altered due to the immaturity of both metabolic enzymes and the gastrointestinal system, which could impact the oral bioavailability of drugs. For example, the total oral bioavailability of midazolam in preterm neonates is higher, approximately three times that of adults [159]. Gastric emptying differs in pediatric populations due to immaturity and does not approach the adult level until 6 to 8 months of age [160,161]. The influence of immature gastric emptying on the absorption rate has yielded conflicting results [161]. Pancreatic lipase secretion matures a few months after birth; therefore, the absorption of fat-soluble drugs is reduced in neonates and young children. In addition, the maturation of transporters should also be assessed when investigating DDIs for CBD/THC in pediatric populations. At birth, P-gp expression is not at adult levels in the intestine, liver, and blood–brain barrier [162,163].
The distributions of THC, CBD, and their metabolites are also different in pediatric populations. Generally, the fat content is relatively low in younger children [161,164]. The fat percentage of body mass peaks around six months of age and then drops to the level that it was at birth during the first ten years [164,165]. The high lipophilicity of THC makes it more sensitive to changes in fat percentage. Plasma protein binding matures in the early stages of life. For example, Maharaj et al. established a nonlinear model to describe the development of α-1-acid glycoprotein protein binding in pediatrics [166]. Additionally, the blood–brain barrier does not fully function after birth, which may result in a higher CBD/THC exposure in the brain [167].
Besides their absorption and distribution, the metabolism of THC, CBD, and their metabolites may also change in pediatrics. CYPs in the phase I metabolism of CBD and THC and UGTs in the phase II metabolism of CBD change rapidly after birth. For example, the CYP3A isoform activity varies in different development stages. The CYP3A4 activity in infants is higher than that in adults and then declines to the adult level. CYP3A4 protein increases rapidly after birth, from low levels to 50% of adult levels by 6 to 12 months of age [168]. At birth, the CYP2C9 activity levels are approximately 30% of the mature values. CYP2C9 protein increases rapidly from birth to five months of age, with half the population having CYP2C9 levels that are comparable to those of adults at five months. CYP2C9 concentrations and activity are relatively stable from 5 months to 18 years old [169]. The CYP2C19 activity is around 15% of the adult level at birth, increasing linearly in the first five months [169,170]. From five months to ten years, the CYP2C19 expression varied 21-fold, and the CYP2C19 concentration and activity level were comparable with adults after ten years of age. Like the phase I metabolism, the phase II metabolism changes during growth. It takes three months to three years or more to increase the activity of hepatic UGTs from <1.0% of adult levels in fetal to approach adult levels [161,162]. The metabolisms of 11-OH-THC, THC-COOH, CBD, 7-OH-CBD, and 7-COOH-CBD may vary at different stages of child growth due to the development of the UGT system, and the changes in the metabolism of the different compounds may also be varied at each growth stage.

3.2.3. Drug Interactions and Their Impact on Pediatric Populations

Only a few studies have investigated DDIs in pediatric populations. Geffrey et al. found that when administered concomitantly with 25 mg/kg/day of CBD, clobazam concentrations increased by over 60%, and N-desmethylclobazam increased by 500% in 25 patients with refractory epilepsy aged 4–19 years [53]. Wheless et al. detected the DDI between CBD and clobazam in 61 pediatric patients aged 1 to 17 years. The CBD concentration increased by 2.5-fold as the victim. When CBD acted as the perpetrator, individuals taking a 40 mg/kg/day CBD oral solution exhibited 1.7- and 2.2-fold higher clobazam concentrations than those taking 10 mg/kg/day and 20 mg/kg/day cannabidiol oral solutions, respectively [52]. Gaston et al. discovered that CBD has DDIs when used concomitantly with topiramate, rufinamide, clobazam, and N-desmethylclobazam. Statistically significant (p < 0.01) increases in the concentrations of topiramate, rufinamide, and N-desmethylclobazam and a decrease in the concentrations of clobazam were observed with increasing the CBD dose from 5 to 50 mg/kg/day in 42 children. They also compared the differences in DDIs between adults and pediatrics, suggesting that DDIs with zonisamide and eslicarbazepine were only observed in adults [51]. A possible reason for this observation is that plasma zonisamide concentrations increase linearly with age, potentially influencing the DDI results [171].
Furthermore, Vaughn et al. used compartment PK models in the software to predict the DDI between serotonin reuptake inhibitors, escitalopram or sertraline, and cannabinoids in adolescents [172]. Their simulations suggested significant increases in both AUC0–24h and Cmax and increased risks of adverse effects after co-administration. When used with concurrent THC or low-dose CBD (5–15 mg/day), escitalopram’s AUC0–24h and Cmax increased by 35% and 25%, respectively, in simulation. Similarly, sertraline’s AUC0–24h and Cmax increased by 33% and 36%, respectively, in simulation.

3.3. Older Adults

3.3.1. Overview of Studies Exploring CBD/THC in Older Adult Populations

Researchers have conducted investigations into the treatment effects of CBD/THC for age-related diseases such as Alzheimer’s disease [173,174,175,176,177], dementia [178,179], and Parkinson’s disease [180,181,182,183,184]. However, most of these studies either focus on the PD of CBD/THC and the clinical endpoint of the disease or combine all age groups when analyzing the PK results. Although the importance of clinical trials in older adults has been recognized since 1979 [118,179,185,186,187], to the best of our knowledge, only Ahmed et al. investigated the PK of THC in older adults in two studies, one in healthy older adults and the other involving older adults with dementia [123,124]. In the randomized controlled trial with healthy older adults, highly variable plasma concentrations of THC, 11-OH-THC, and THC-COOH were reported. The THC AUC02h (n = 11) ranged from 1.67 to 3.51 ng∙h/mL, and the Cmax was 1.42 to 4.57 ng/mL for those within the two-hour sampling period (Table 3). The Tmax of the THC of some older adults was not observed during the 120 min sampling periods, whereas the Tmax of the younger adults mainly were within 60 min with the same THC dosage form. Notably, in this study, the final samples were collected 2 h post-dose. Due to the long half-life of THC, the PK profiles of THC were incomplete [123]. A similar delayed Tmax (120–180 min) was also observed in trials with older patients with dementia. Additionally, the AUC06h of older adults with dementia was two times higher than that of the younger adults, with a CV of up to 136% being observed (Table 3) [124]. It should be noted that in the study of older adults with dementia, THC was taken with food, while in the study of younger adults, THC was taken during fasting. Since food increases both the AUC and Cmax of THC, the conclusion that AUC is increased in older adults with dementia may be biased.

3.3.2. Age-Related Changes in PK and Implications for CBD/THC

Aging has an impact on every aspect of CBD/THC PK, from absorption to excretion. The stomach, intestine, and liver play the most important roles in the absorption of CBD and THC when they are administered orally; due to the high lipophilicity of CBD/THC, the pancreas may also influence their absorption. Lungs and the oral cavity can also affect absorption when inhaled, administered sublingually, or by means of an oromucosal spray. The possible characteristics of an aged stomach could be hypochlorhydria and a decreased volume of gastrointestinal fluids [188,189]. Although most in vivo studies suggested that the gastric emptying time increased in older adults [190,191,192], a conflicting study showed that the emptying rate of older adults remains within the same range as that of young adults [193]. The hypochlorhydria may decrease CBD/THC dissolution and thus change the systemic exposure, especially for the powder and capsules [194]. The volume of gastric and intestinal fluids decreases in older adults [195,196]. Since self-emulsifying drug delivery systems (SEDDSs) require water or gastrointestinal fluid for emulsification, their absorption may be delayed. In older adults, the blood flow in the intestine and the small bowel surface decreases, impairing absorption. The changes in the stomach and intestine may explain the delayed Tmax observed in the two clinical trials of Ahmed et al. [123,124].
Due to its low solubility in water, CBD/THC is often given with oil or SEDDS to increase its bioavailability. Pancreatic lipases hydrolyze the lipids in the oil, and the SEDDS oil phases in the intestine [197], and a decrease in pancreatic lipase secretion due to senescence may eliminate the bioavailability advantage of oil formulations and SEDDSs [198]. For an inhalation formulation, the effect of aging on absorption varies. Although the overall lung function decreased in older adults, physiological changes such as an increased alveolar–capillary surface area, alveolar size, end-expiratory lung volume, and functional residual capacity, along with a reduced expiratory airflow, make the influence of the declined lung function on the trans-pulmonary absorption of CBD/THC uncertain [199,200,201]. Salivary gland secretion decreased with aging, and the quality of saliva changed, resulting in reduced absorption of sublingual formulations and oromucosal sprays through the oral cavity [202,203].
Changes in fat composition, cardiovascular senescence, and the decrease in plasma protein are the main factors affecting drug distribution during aging. Generally, the total fat mass increases in older adults, and the fat redistribution to peripheral tissues (e.g., abdominal fat) increases [204]. The volume of distributions of lipid-soluble CBD/THC and their metabolites may increase. Cardiovascular senescence could lead to a decline in cardiac output and heart failure [205], which mediates hypoperfusion in the peripheral tissues and delays the distribution of CBD/THC to the periphery [206]. Due to brain diseases in older adults, the permeability of the blood–brain barrier may increase, therefore increasing the distribution of CBD/THC in the brain [207]. Plasma protein, mainly lipoproteins, followed by albumin, is bound to 95–99% of plasma THC [104,105]. The change in lipoproteins and albumin may impact the concentration of CBD/THC. A 20% increase in lipoprotein levels was observed in older adults over 60 years old. Serum albumin levels decreased by 20% at age 70 [208], while there appears to be very little change in α1-acid glycoprotein (AAG) concentrations in older adults [209].
The liver impacts the metabolism of CBD and THC. Both the liver’s blood flow and volume decrease with aging. The liver may lose 20–40% of its volume and around 40–60% of its blood flow in older adults compared with younger adults [210,211]. In addition to the physiological change in the liver, senescence may affect the functions of drug-metabolizing enzymes [212]. Age-related changes in CYP enzymes have been assessed in various studies using both in vitro and in vivo methods and have yielded some conflicting results [213]. UGTs have not been reported to decrease with aging, but one animal study suggests that UGT1A1 decreases in elderly mice [213]. The liver also impacts absorption through the first-pass metabolism, an important factor in oral bioavailability. With the aforementioned changes in the liver and the high prevalence of fibrosis, the first-pass metabolism may be reduced, resulting in a higher AUC.
THC is excreted mainly as acid metabolites, about 65–80% in feces and 20–35% in urine, and the amount THC being excreted as a parent drug is limited. The lower renal metabolite excretion can be explained by tubular reabsorption [4]. Like the liver, the kidney’s size, weight, and renal blood flow decreased in older adults. The kidney loses one-fourth of its weight in older adults, and the renal blood flow decreases by 10% per decade, starting at 40 years old. Moreover, the estimated glomerular filtration rate and renal tubular reabsorption capacity decreased [214]. The overall effect of aging on the renal excretion of CBD and THC and their metabolites is unknown.

3.3.3. Drug Interactions and Considerations for Older Adult Populations

In addition to the growing use of recreational cannabis in older adults, the potential medical use of cannabis by older adults is also on the rise for the treatment of pain, insomnia, anxiety, depression, and Parkinson’s disease [215,216]. On the other hand, polypharmacy is often encountered in older adults, which makes the DDI considerations particularly important in older adults, especially considering that CBD/THC could be either a perpetrator or victim in DDIs. However, there is a lack of data to substantiate either of these potential drug interactions in older adults. Some case reports investigated potential DDIs between CBD/THC and medications such as warfarin (85-year-old male) and clopidogrel that was co-administered with aspirin (76-year-old male) in older adults [71,97]. The international normalized ratio (INR) was unchanged in the case report of an 85-year-old male patient using warfarin. In contrast, five case reports in young adults revealed INR elevations in patients on warfarin receiving cannabis. However, the dose of CBD and THC was relatively low in the case report of the older adult, which was 5.3 mg CBD and 0.3 mg of THC once daily [97], compared with a daily dose of 10.3 mg CBD and 14.7 mg THC reported in the case report of DDI effects in young adults [98]. Brown et al. reported a case where the co-medication of CBD with clopidogrel and aspirin resulted in acute myocardial infarction in a 76-year-old man [71]. More clinical studies are essential to assess the DDIs between CBD/THC and other drugs in older adults.

3.4. Patients with Hepatic or Renal Impairment

3.4.1. Overview of Studies Examining CBD/THC in Patients with Hepatic or Renal Impairment

PK studies of CBD/THC in patients with hepatic or renal impairment are limited. Interestingly, Suzuki et al. suggested that CBD demonstrated a treatment effect on renal fibrosis, significantly reduced apoptosis in the kidney, and partially protected the kidney from cisplatin-induced nephrotoxicity. The proposed mechanisms of the renoprotection effects are suppressing the mRNA of inflammatory cytokines in cisplatin-induced nephropathy and reducing apoptosis by inhibiting caspase-3 activity [217]. Another animal study showed that CBD exhibited significant renoprotection against acute kidney injury by favoring the development of regulatory T-17 (Treg 17) cells, thus inhibiting the proinflammatory responses that lead to kidney injury [218]. Two reviews also investigated the effect of CBD and CBD co-medicated with THC on symptoms associated with kidney failure, demonstrating positive effects on uremic pruritus, restless legs syndrome, anorexia, and nausea. These treatment effects may be due to the actions of CBD and THC on a range of neuronal receptors and ion channels, including CB1 receptors, CB2 receptors, gamma-aminobutyric acid A receptors, alpha-3 glycine receptors, 5HT1a receptors, transient receptor potential vanilloid type 1 (TRPV1), TRPV2, TRPV3, and TRPV4 [219,220]. For hepatic impairment, a study found that CB2 agonists reduced liver impairment and accelerated liver regeneration in mice [221].

3.4.2. Impact of Hepatic Impairment on CBD/THC PK

Taylor et al. assessed the PK of a single oral dose of 200 mg CBD solution in healthy volunteers and patients with mild-to-severe hepatic impairment. The PK parameters of CBD in this trial are presented in Table 2. The results suggested that the AUC0–t and Cmax of CBD and its metabolites increase with the severity of hepatic impairment, except for 7-COOH-CBD in patients with severe hepatic impairment. The AUC0–t and Cmax were lower in patients with severe hepatic impairment than in healthy volunteers. The AUC0–t of CBD was 44.3%, 134.7%, and 313.1% higher in patients with mild, moderate, and severe hepatic impairment, respectively, compared with volunteers with normal hepatic function. Similarly, the Cmax of CBD was 57.4%, 139.2%, and 159.5% higher, respectively [117]. Bansal et al. developed and verified the effect of hepatic impairment on CBD by using PBPK modeling. In simulation, they captured the AUC and Cmax change in patients with mild, moderate, and severe hepatic impairment [158]. To date, no clinical trials have reported the PK of THC in patients with hepatic impairment. Current prescribing information for Epidiolex® recommends an initial dose adjustment and slower dose titration for patients with moderate-to-severe hepatic impairment [15].

3.4.3. Impact of Renal Impairment on CBD/THC PK

Tayo et al. investigated the PK of a single dose of 200 mg CBD oral solution in subjects ranging from normal renal function to severe renal impairment, and the PK parameters of CBD in renal impairment patients are listed in Table 2. They suggested that the exposure to CBD and its major metabolites did not differ between healthy volunteers and patients with any degree of renal impairment. The AUC0–t of CBD was 44%, 14%, and 15% higher in patients with mild, moderate, and severe renal impairment, respectively, compared with the normal renal function group. It is noted that the mild renal impairment group had the lowest mean body mass index, which may lead to an increase in CBD’s AUC0–t. According to their results, no dose adjustment of CBD is required in patients with renal impairment [118]. To date, no clinical trials have investigated the PK of THC in patients with renal impairment.

3.4.4. Drug Interactions and Considerations for Patients with Hepatic or Renal Impairment

Current research findings show no effect of renal impairment on the PK of CBD. Thus, drugs that affect renal function may have no impact on CBD, and no dose adjustment is needed for CBD in patients with renal impairment. To date, no clinical trials have reported the PK of THC in patients with hepatic or renal impairment. More clinical trials may be required to characterize the impact of DDIs in patients with hepatic and renal impairment.

3.5. Other Special Populations

3.5.1. CBD/THC Studies in Cancer Patients

Most of the CBD/THC studies in cancer patients focus on their PD effects, and only one pilot clinical study reported the PK of CBD and THC in patients with cancer. They found a large variability of both CBD and THC in cancer patients. The AUC0–t of THC and CBD ranged from 1.11 to 6.61 ng∙h/mL and 0.49 to 4.06 ng∙h/mL, respectively, after a single dose of 2.5 mg THC and 2.5 mg CBD. Additionally, the t1/2 of CBD and THC increased with the dose. The median t1/2 for 2.5 mg of CBD was 0.75 h, while the median t1/2 for 7.5 mg of CBD was 1.53 h. Similarly, for 2.5 mg of THC, the t1/2 was 0.94 h, whereas for 7.5 mg of THC, the t1/2 was 1.39 h (Table 2 and Table 3) [119]. The effects of CBD/THC in cancer patients primarily focus on reducing symptom distress, including cancer-related pain, cachexia, nausea, and vomiting. However, conflicting results have been reported in clinical trials, and findings may lack statistical power [222,223,224,225]. A recent retrospective chart review suggested that there was no statistically significant change in symptom distress with medical cannabis use in cancer patients [226,227]. In contrast, some studies report that CBD/THC may be effective in cancer-related neuropathic pain and in pain that is inadequately controlled by opioid therapy [228]. Notably, a phase Ib trial showed that the CBD and THC complex has the potential to improve the one-year survival in glioblastoma patients, while no DDI effect of CBD and THC on temozolomide PK was observed [186].

3.5.2. CBD/THC Studies in Patients with Neurological Disorders

Various clinical studies have been conducted with CBD as a potential treatment for neuropsychiatric disorders, including epilepsy, movement disorders, neuropathic pain, anxiety, schizophrenia, and substance abuse disorder; however, small sample sizes, conflicting results, and variability in study designs make determining its efficacy difficult [229,230,231,232,233]. The most rigorously tested and only FDA-approved indication for CBD is for the treatment of seizures associated with Lennox–Gastaut syndrome, Dravet syndrome, and tuberous sclerosis complex. Gherzi et al. reported the PK of CBD and THC in young patients with drug-resistant epilepsy [234]. Sativex® has also been approved outside the US as an adjunctive treatment for spasticity related to multiple sclerosis [235]. A recent systematic review of randomized controlled trials comparing cannabinoids with conventional treatments or placebo found moderate evidence for analgesic benefit in patients with chronic neuropathic pain [236]. Additional studies have suggested that CBD may also be effective in treating symptoms of opioid addiction [237,238].

3.5.3. CBD/THC Studies in Other Special Populations

In addition to the populations mentioned above, some research investigated the use of CBD/THC in several other populations. CBD and THC were reported to have the potential to benefit people with inflammatory bowel diseases [239,240]; the PK parameters of CBD and THC in Crohn’s disease are listed in Table 2 and Table 3. CBD also reduces pancreatic inflammation caused by immune cell invasion in mice, which can lead to type 1 diabetes [241]. On the other hand, a phase II study suggested that CBD has the potential to decrease the incidence of acute graft-versus-host disease [242]. Populations with HIV are another relative hotspot in CBD/THC studies. The effect of cannabis on the viral load in HIV-infected patients was investigated, and daily/near-daily cannabis use was associated with HIV viral load suppression [243,244]. Some studies also suggested that cannabis can improve pain and other medical symptoms of HIV [245,246,247,248]. Finally, cannabis was reported to enhance the global cognitive performance in older adult patients with HIV [249].

4. Conclusions

4.1. Summary of Key Findings on CBD/THC PK and Drug Interactions

The PK of CBD and THC have been characterized in several studies, but research gaps remain. Large inter-individual variability in PK has been observed in various publications, the reasons for which have not been well investigated. Due to the lack of research, there is also a knowledge gap about PK in special populations. DDIs between CBD/THC and a few therapeutic drugs, such as antiepileptic drugs, have been investigated in clinical trials, but more potential DDIs remain to be explored in clinical trials. As hemp-based CBD is legal at a federal level in the US and some CBD products contain THC, more special populations may be exposed to CBD and THC. Additionally, the absorption, distribution, and metabolism may differ in special populations, as mentioned in the section above. This emphasizes the necessity of evaluating potential DDIs between CBD/THC and medications that are commonly used in these populations.

4.2. Implications for Special Populations and Other Considerations

While well-designed clinical trials remain the gold standard for assessing PK and DDIs in special populations, challenges such as difficulty in recruitment, polypharmacy, individual genetic variation, and treatment compliance have hindered the conduct of such studies. The modeling approaches, particularly PBPK modeling, have become a method for predicting the PK in special populations and determine the potential DDIs. PBPK models incorporate systems biology to provide a more mechanistic approach for analyzing populations that are difficult to represent adequately. Some research groups have developed PBPK models for CBD or THC, as covered in the sections where the PK of CBD were investigated by modeling in special populations, including adults with hepatic impairment and children, while the PK of THC were also studied in pregnant and lactating women.

4.3. Future Research Directions and Areas of Further Investigation

However, PBPK relies on our knowledge of the target population’s physiology and the drug’s properties. The absorption of different CBD and THC formulations has not been well described by the established models yet. Moreover, the roles and contribution of CYPs and UGTs in the metabolism of CBD and THC and their metabolites vary widely among studies. In addition, there are conflicting findings regarding the physiology of special populations. Overall, more work is required to comprehensively investigate the PK and DDIs of CBD and THC in special populations. New in vitro and in vivo technologies, such as organ-on-a-chip, microsampling, and microdialysis, may also enhance our physiological understanding and improve model predictions. Utilizing these physiological models in combination with clinical trials and modeling approaches could help better manage DDIs associated with CBD and THC in special populations.

Author Contributions

Writing—original draft preparation, L.Q., J.L.B. and Z.Z.; writing—review and editing, K.D.J. and Z.Z.; project administration, Z.Z.; funding acquisition, Z.Z. All authors have read and agreed to the published version of the manuscript.

Funding

Z.Z. was supported by NIH/NIGMS, grant number R16 GM146679 and 2023ASCPT Darrell Abernethy Early Stage Investigator Award. K.D.J. was supported by NIH/NIGMS, award number R35GM143044. J.L.B. was supported by NIH/NIGMS, grant number T32GM086330. This review is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

Data Availability Statement

The data presented in this study are available in this article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Cox, E.J.; Maharao, N.; Patilea-Vrana, G.; Unadkat, J.D.; Rettie, A.E.; McCune, J.S.; Paine, M.F. A Marijuana-Drug Interaction Primer: Precipitants, Pharmacology, and Pharmacokinetics. Pharmacol. Ther. 2019, 201, 25–38. [Google Scholar] [CrossRef]
  2. Lafaye, G.; Karila, L.; Blecha, L.; Benyamina, A. Cannabis, Cannabinoids, and Health. Dialogues Clin. Neurosci. 2017, 19, 309–316. [Google Scholar] [CrossRef]
  3. Zhou, Z.; Paine, M.F.; Spindle, T.R.; Huang, S.; Zhang, L. Cannabis for Medical Use: Clinical Pharmacology Perspectives on Scientific and Regulatory Challenges. Clin. Pharmacol. Ther. 2022, 111, 732–735. [Google Scholar] [CrossRef] [PubMed]
  4. Lucas, C.J.; Galettis, P.; Schneider, J. The Pharmacokinetics and the Pharmacodynamics of Cannabinoids. Br. J. Clin. Pharmacol. 2018, 84, 2477–2482. [Google Scholar] [CrossRef] [PubMed]
  5. Kesner, A.J.; Lovinger, D.M. Cannabis Use, Abuse, and Withdrawal: Cannabinergic Mechanisms, Clinical, and Preclinical Findings. J. Neurochem. 2021, 157, 1674–1696. [Google Scholar] [CrossRef]
  6. Pertwee, R. The Diverse CB1 and CB2 Receptor Pharmacology of Three Plant Cannabinoids: D9-Tetrahydrocannabinol, Cannabidiol and D9-Tetrahydrocannabivarin. Br. J. Pharmacol. 2008, 153, 199–215. [Google Scholar] [CrossRef] [PubMed]
  7. Oberbarnscheidt, T.; Miller, N.S. The Impact of Cannabidiol on Psychiatric and Medical Conditions. J. Clin. Med. Res. 2020, 12, 393–403. [Google Scholar] [CrossRef]
  8. Tham, M.; Yilmaz, O.; Alaverdashvili, M.; Kelly, M.E.M.; Denovan-Wright, E.M.; Laprairie, R.B. Allosteric and Orthosteric Pharmacology of Cannabidiol and Cannabidiol-dimethylheptyl at the Type 1 and Type 2 Cannabinoid Receptors. Br. J. Pharmacol. 2019, 176, 1455–1469. [Google Scholar] [CrossRef]
  9. Chung, H.; Fierro, A.; Pessoa-Mahana, C.D. Cannabidiol Binding and Negative Allosteric Modulation at the Cannabinoid Type 1 Receptor in the Presence of Delta-9-Tetrahydrocannabinol: An In Silico Study. PLoS ONE 2019, 14, e0220025. [Google Scholar] [CrossRef]
  10. Britch, S.C.; Babalonis, S.; Walsh, S.L. Cannabidiol: Pharmacology and Therapeutic Targets. Psychopharmacology 2021, 238, 9–28. [Google Scholar] [CrossRef]
  11. Story, G.; Briere, C.-E.; McClements, D.J.; Sela, D.A. Cannabidiol and Intestinal Motility: A Systematic Review. Curr. Dev. Nutr. 2023, 7, 101972. [Google Scholar] [CrossRef] [PubMed]
  12. Smith, T.; Majid, F.; Eckl, V.; Morton Reynolds, C. Herbal Supplement Sales in US Increase by Record-Breaking 17.3% in 2020. Available online: https://www.herbalgram.org/resources/herbalgram/issues/131/table-of-contents/hg131-mkrpt/ (accessed on 31 January 2024).
  13. Smith, T.; Resetar, H.; Morton, C. US Sales of Herbal Supplements Increase by 9.7% in 2020; Herbalgram Report; American Botanical Council: Austin, TX, USA., 2021. [Google Scholar]
  14. Jazz Pharmaceuticals EPIDIOLEX (Cannabidiol) Prescribing Information. Available online: https://pp.jazzpharma.com/pi/epidiolex.en.USPI.pdf (accessed on 12 November 2023).
  15. Jazz Pharmaceuticals Epidyolex (Cannabidiol) Product Information. Available online: https://www.ema.europa.eu/en/documents/product-information/epidyolex-epar-product-information_en.pdf (accessed on 12 November 2023).
  16. Nichols, J.M.; Kaplan, B.L.F. Immune Responses Regulated by Cannabidiol. Cannabis Cannabinoid Res. 2020, 5, 12–31. [Google Scholar] [CrossRef] [PubMed]
  17. Esposito, G.; Scuderi, C.; Valenza, M.; Togna, G.I.; Latina, V.; De Filippis, D.; Cipriano, M.; Carratù, M.R.; Iuvone, T.; Steardo, L. Cannabidiol Reduces Aβ-Induced Neuroinflammation and Promotes Hippocampal Neurogenesis through PPARγ Involvement. PLoS ONE 2011, 6, e28668. [Google Scholar] [CrossRef] [PubMed]
  18. De Filippis, D.; Esposito, G.; Cirillo, C.; Cipriano, M.; De Winter, B.Y.; Scuderi, C.; Sarnelli, G.; Cuomo, R.; Steardo, L.; De Man, J.G.; et al. Cannabidiol Reduces Intestinal Inflammation through the Control of Neuroimmune Axis. PLoS ONE 2011, 6, e28159. [Google Scholar] [CrossRef] [PubMed]
  19. Chandra, S.; Radwan, M.M.; Majumdar, C.G.; Church, J.C.; Freeman, T.P.; ElSohly, M.A. New Trends in Cannabis Potency in USA and Europe during the Last Decade (2008–2017). Eur. Arch. Psychiatry Clin. Neurosci. 2019, 269, 5–15. [Google Scholar] [CrossRef]
  20. Alkem Labs. Marinol (Dronabinol) [Package Insert]. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2006/018651s025s026lbl.pdf (accessed on 2 November 2023).
  21. Benuvia. Syndros (Dronabinol) [Package Insert]. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/205525s003lbl.pdf (accessed on 2 November 2023).
  22. Jazz Pharmaceuticals, Inc. Sativex (Delta-9-Tetrahydrocannabinol and Cannabidiol) Oromucosal Spray Electronic Medicines Compendium. Available online: https://www.medicines.org.uk/emc/product/602/ (accessed on 12 November 2023).
  23. Jazz Pharmaceuticals, Inc. Sativex (Delta-9-Tetrahydrocannabinol and Cannabidiol) Oromucosal Spray. Available online: https://www.jazzpharma.com/medicines/our-medicines/ (accessed on 12 November 2023).
  24. Brown, J.D.; Winterstein, A.G. Potential Adverse Drug Events and Drug-Drug Interactions with Medical and Consumer Cannabidiol (CBD) Use. J. Clin. Med. 2019, 8, 989. [Google Scholar] [CrossRef] [PubMed]
  25. Miller, I.; Scheffer, I.E.; Gunning, B.; Sanchez-Carpintero, R.; Gil-Nagel, A.; Perry, M.S.; Saneto, R.P.; Checketts, D.; Dunayevich, E.; Knappertz, V. Dose-Ranging Effect of Adjunctive Oral Cannabidiol vs Placebo on Convulsive Seizure Frequency in Dravet Syndrome: A Randomized Clinical Trial. JAMA Neurol. 2020, 77, 613–621. [Google Scholar] [CrossRef] [PubMed]
  26. Petrilli, K.; Ofori, S.; Hines, L.; Taylor, G.; Adams, S.; Freeman, T.P. Association of Cannabis Potency with Mental Ill Health and Addiction: A Systematic Review. Lancet Psychiatry 2022, 9, 736–750. [Google Scholar] [CrossRef]
  27. Barrus, D.G.; Capogrossi, K.L.; Cates, S.C.; Gourdet, C.K.; Peiper, N.C.; Novak, S.P.; Lefever, T.W.; Wiley, J.L. Tasty THC: Promises and Challenges of Cannabis Edibles. Methods Rep. RTI Press 2016, 2016. [Google Scholar] [CrossRef]
  28. Hines, L.A.; Freeman, T.P.; Gage, S.H.; Zammit, S.; Hickman, M.; Cannon, M.; Munafo, M.; MacLeod, J.; Heron, J. Association of High-Potency Cannabis Use with Mental Health and Substance Use in Adolescence. JAMA Psychiatry 2020, 77, 1044–1051. [Google Scholar] [CrossRef]
  29. Pichini, S.; Mannocchi, G.; Berretta, P.; Zaami, S.; Pirani, F.; Pacifici, R.; Busardò, F.P. Δ9-Tetrahydrocannabinol and Cannabidiol Time Courses in the Sera of “Light Cannabis” Smokers: Discriminating Light Cannabis Use from Illegal and Medical Cannabis Use. Ther. Drug Monit. 2020, 42, 151–156. [Google Scholar] [CrossRef] [PubMed]
  30. Busardò, F.P.; Pérez-Acevedo, A.P.; Pacifici, R.; Mannocchi, G.; Gottardi, M.; Papaseit, E.; Pérez-Mañá, C.; Martin, S.; Poyatos, L.; Pichini, S.; et al. Disposition of Phytocannabinoids, Their Acidic Precursors and Their Metabolites in Biological Matrices of Healthy Individuals Treated with Vaporized Medical Cannabis. Pharmaceuticals 2021, 14, 59. [Google Scholar] [CrossRef] [PubMed]
  31. Pérez-Acevedo, A.P.; Pacifici, R.; Mannocchi, G.; Gottardi, M.; Poyatos, L.; Papaseit, E.; Pérez-Mañá, C.; Martin, S.; Busardò, F.P.; Pichini, S.; et al. Disposition of Cannabinoids and Their Metabolites in Serum, Oral Fluid, Sweat Patch and Urine from Healthy Individuals Treated with Pharmaceutical Preparations of Medical Cannabis. Phytother. Res. 2021, 35, 1646–1657. [Google Scholar] [CrossRef] [PubMed]
  32. Stella, B.; Baratta, F.; Della Pepa, C.; Arpicco, S.; Gastaldi, D.; Dosio, F. Cannabinoid Formulations and Delivery Systems: Current and Future Options to Treat Pain. Drugs 2021, 81, 1513–1557. [Google Scholar] [CrossRef] [PubMed]
  33. Perucca, E.; Bialer, M. Critical Aspects Affecting Cannabidiol Oral Bioavailability and Metabolic Elimination, and Related Clinical Implications. CNS Drugs 2020, 34, 795–800. [Google Scholar] [CrossRef] [PubMed]
  34. Mozaffari, K.; Willette, S.; Lucker, B.F.; Kovar, S.E.; Holguin, F.O.; Guzman, I. The Effects of Food on Cannabidiol Bioaccessibility. Molecules 2021, 26, 3573. [Google Scholar] [CrossRef]
  35. Martinez Naya, N.; Kelly, J.; Corna, G.; Golino, M.; Polizio, A.H.; Abbate, A.; Toldo, S.; Mezzaroma, E. An Overview of Cannabidiol as a Multifunctional Drug: Pharmacokinetics and Cellular Effects. Molecules 2024, 29, 473. [Google Scholar] [CrossRef] [PubMed]
  36. Yeung, C.H.T.; Beers, J.L.; Jackson, K.D.; Edginton, A.N. Verifying in Vitro-determined Enzyme Contributions to Cannabidiol Clearance for Exposure Predictions in Human through Physiologically-based Pharmacokinetic Modeling. CPT Pharmacomet. Syst. Pharmacol. 2023, 12, 320–332. [Google Scholar] [CrossRef]
  37. 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, 165–170. [Google Scholar] [CrossRef]
  38. Mazur, A.; Lichti, C.F.; Prather, P.L.; Zielinska, A.K.; Bratton, S.M.; Gallus-Zawada, A.; Finel, M.; Miller, G.P.; Radomińska-Pandya, A.; Moran, J.H. Characterization of Human Hepatic and Extrahepatic UDP-Glucuronosyltransferase Enzymes Involved in the Metabolism of Classic Cannabinoids. Drug Metab. Dispos. 2009, 37, 1496–1504. [Google Scholar] [CrossRef]
  39. Huestis, M.A. Human Cannabinoid Pharmacokinetics. Chem. Biodivers. 2007, 4, 1770–1804. [Google Scholar] [CrossRef]
  40. Huestis, M.A.; Sempio, C.; Newmeyer, M.N.; Andersson, M.; Barnes, A.J.; Abulseoud, O.A.; Blount, B.C.; Schroeder, J.; Smith, M.L. Free and Glucuronide Urine Cannabinoids after Controlled Smoked, Vaporized and Oral Cannabis Administration in Frequent and Occasional Cannabis Users. J. Anal. Toxicol. 2020, 44, 651–660. [Google Scholar] [CrossRef] [PubMed]
  41. Bardhi, K.; Coates, S.; Watson, C.J.W.; Lazarus, P. Cannabinoids and Drug Metabolizing Enzymes: Potential for Drug-Drug Interactions and Implications for Drug Safety and Efficacy. Expert. Rev. Clin. Pharmacol. 2022, 15, 1443–1460. [Google Scholar] [CrossRef] [PubMed]
  42. Matheson, J.; Sproule, B.; Di Ciano, P.; Fares, A.; Le Foll, B.; Mann, R.E.; Brands, B. Sex Differences in the Acute Effects of Smoked Cannabis: Evidence from a Human Laboratory Study of Young Adults. Psychopharmacology 2020, 237, 305–316. [Google Scholar] [CrossRef] [PubMed]
  43. Sallam, N.A.; Peterson, C.S.; Baglot, S.L.; Kohro, Y.; Trang, T.; Hill, M.N.; Borgland, S.L. Sex Differences in Plasma, Adipose Tissue, and Central Accumulation of Cannabinoids, and Behavioral Effects of Oral Cannabis Consumption in Male and Female C57BL/6 Mice. Int. J. Neuropsychopharmacol. 2023, 26, pyad055. [Google Scholar] [CrossRef] [PubMed]
  44. Millar, S.A.; Stone, N.L.; Yates, A.S.; O’Sullivan, S.E. A Systematic Review on the Pharmacokinetics of Cannabidiol in Humans. Front. Pharmacol. 2018, 9, 1365. [Google Scholar] [CrossRef] [PubMed]
  45. Alves, P.; Amaral, C.; Teixeira, N.; Correia-da-Silva, G. Cannabis Sativa: Much More beyond Δ9-Tetrahydrocannabinol. Pharmacol. Res. 2020, 157, 104822. [Google Scholar] [CrossRef]
  46. Kebede, L.; Masoomi Dezfooli, S.; Seyfoddin, A. Medicinal Cannabis Pharmacokinetics and Potential Methods of Delivery. Pharm. Dev. Technol. 2022, 27, 202–214. [Google Scholar] [CrossRef] [PubMed]
  47. Huestis, M.A. Pharmacokinetics and Metabolism of the Plant Cannabinoids, Δ9-Tetrahydrocannabinol, Cannabidiol and Cannabinol. In Cannabinoids; Handbook of Experimental Pharmacology; Springer: Berlin/Heidelberg, Germany, 2005; pp. 657–690. [Google Scholar] [CrossRef]
  48. 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. Pharm. Drug Dev. 2019, 8, 1009–1031. [Google Scholar] [CrossRef]
  49. VanLandingham, K.E.; Crockett, J.; Taylor, L.; Morrison, G. A Phase 2, Double-Blind, Placebo-Controlled Trial to Investigate Potential Drug-Drug Interactions between Cannabidiol and Clobazam. J. Clin. Pharmacol. 2020, 60, 1304–1313. [Google Scholar] [CrossRef]
  50. Ben-Menachem, E.; Gunning, B.; Arenas Cabrera, C.M.; VanLandingham, K.; Crockett, J.; Critchley, D.; Wray, L.; Tayo, B.; Morrison, G.; Toledo, M. A Phase II Randomized Trial to Explore the Potential for Pharmacokinetic Drug–Drug Interactions with Stiripentol or Valproate When Combined with Cannabidiol in Patients with Epilepsy. CNS Drugs 2020, 34, 661–672. [Google Scholar] [CrossRef] [PubMed]
  51. Gaston, T.E.; Bebin, E.M.; Cutter, G.R.; Liu, Y.; Szaflarski, J.P.; the UAB CBD Program. Interactions between Cannabidiol and Commonly Used Antiepileptic Drugs. Epilepsia 2017, 58, 1586–1592. [Google Scholar] [CrossRef]
  52. Wheless, J.W.; Dlugos, D.; Miller, I.; Oh, D.A.; Parikh, N.; Phillips, S.; Renfroe, J.B.; Roberts, C.M.; Saeed, I.; Sparagana, S.P.; et al. Pharmacokinetics and Tolerability of Multiple Doses of Pharmaceutical-Grade Synthetic Cannabidiol in Pediatric Patients with Treatment-Resistant Epilepsy. CNS Drugs 2019, 33, 593–604. [Google Scholar] [CrossRef] [PubMed]
  53. Geffrey, A.L.; Pollack, S.F.; Bruno, P.L.; Thiele, E.A. Drug-Drug Interaction between Clobazam and Cannabidiol in Children with Refractory Epilepsy. Epilepsia 2015, 56, 1246–1251. [Google Scholar] [CrossRef] [PubMed]
  54. Tanwir, A.; Szabó, C.Á. Non-Convulsive Status Epilepticus in the Setting of Cannabidiol Adjunctive Therapy. Epileptic Disord. 2022, 24, 713–718. [Google Scholar] [CrossRef] [PubMed]
  55. Cortopassi, J. Warfarin Dose Adjustment Required after Cannabidiol Initiation and Titration. Am. J. Health Syst. Pharm. 2020, 77, 1846–1851. [Google Scholar] [CrossRef] [PubMed]
  56. Damkier, P.; Lassen, D.; Christensen, M.M.H.; Madsen, K.G.; Hellfritzsch, M.; Pottegård, A. Interaction between Warfarin and Cannabis. Basic. Clin. Pharmacol. Toxicol. 2019, 124, 28–31. [Google Scholar] [CrossRef] [PubMed]
  57. Grayson, L.; Vines, B.; Nichol, K.; Szaflarski, J.P.; UAB CBD Program. An Interaction between Warfarin and Cannabidiol, a Case Report. Epilepsy Behav. Case Rep. 2018, 9, 10–11. [Google Scholar] [CrossRef] [PubMed]
  58. Hsu, A.; Painter, N.A. Probable Interaction between Warfarin and Inhaled and Oral Administration of Cannabis. J. Pharm. Pract. 2020, 33, 915–918. [Google Scholar] [CrossRef]
  59. Yamreudeewong, W.; Wong, H.K.; Brausch, L.M.; Pulley, K.R. Probable Interaction between Warfarin and Marijuana Smoking. Ann. Pharmacother. 2009, 43, 1347–1353. [Google Scholar] [CrossRef]
  60. Zullino, D.F.; Delessert, D.; Eap, C.B.; Preisig, M.; Baumann, P. Tobacco and Cannabis Smoking Cessation Can Lead to Intoxication with Clozapine or Olanzapine. Int. Clin. Psychopharmacol. 2002, 17, 141–143. [Google Scholar] [CrossRef] [PubMed]
  61. Vierke, C.; Marxen, B.; Boettcher, M.; Hiemke, C.; Havemann-Reinecke, U. Buprenorphine-Cannabis Interaction in Patients Undergoing Opioid Maintenance Therapy. Eur. Arch. Psychiatry Clin. Neurosci. 2021, 271, 847–856. [Google Scholar] [CrossRef] [PubMed]
  62. Leino, A.D.; Emoto, C.; Fukuda, T.; Privitera, M.; Vinks, A.A.; Alloway, R.R. Evidence of a Clinically Significant Drug-Drug Interaction between Cannabidiol and Tacrolimus. Am. J. Transpl. 2019, 19, 2944–2948. [Google Scholar] [CrossRef] [PubMed]
  63. Ebrahimi-Fakhari, D.; Agricola, K.D.; Tudor, C.; Krueger, D.; Franz, D.N. Cannabidiol Elevates Mechanistic Target of Rapamycin Inhibitor Levels in Patients with Tuberous Sclerosis Complex. Pediatr. Neurol. 2020, 105, 59–61. [Google Scholar] [CrossRef] [PubMed]
  64. Wiemer-Kruel, A.; Stiller, B.; Bast, T. Cannabidiol Interacts Significantly with Everolimus—Report of a Patient with Tuberous Sclerosis Complex. Neuropediatrics 2019, 50, 400–403. [Google Scholar] [CrossRef]
  65. Thai, C.; Tayo, B.; Critchley, D. A Phase 1 Open-Label, Fixed-Sequence Pharmacokinetic Drug Interaction Trial to Investigate the Effect of Cannabidiol on the CYP1A2 Probe Caffeine in Healthy Subjects. Clin. Pharmacol. Drug Dev. 2021, 10, 1279–1289. [Google Scholar] [CrossRef] [PubMed]
  66. Anderson, L.L.; Doohan, P.T.; Oldfield, L.; Kevin, R.C.; Arnold, J.C.; Berger, M.; Amminger, G.P.; McGregor, I.S. Citalopram and Cannabidiol: In Vitro and In Vivo Evidence of Pharmacokinetic Interactions Relevant to the Treatment of Anxiety Disorders in Young People. J. Clin. Psychopharmacol. 2021, 41, 525–533. [Google Scholar] [CrossRef]
  67. Jusko, W.J.; Gardner, M.J.; Mangione, A.; Schentag, J.J.; Koup, J.R.; Vance, J.W. Factors Affecting Theophylline Clearances: Age, Tobacco, Marijuana, Cirrhosis, Congestive Heart Failure, Obesity, Oral Contraceptives, Benzodiazepines, Barbiturates, and Ethanol. J. Pharm. Sci. 1979, 68, 1358–1366. [Google Scholar] [CrossRef] [PubMed]
  68. Jusko, W.J.; Schentag, J.J.; Clark, J.H.; Gardner, M.; Yurchak, A.M. Enhanced Biotransformation of Theophylline in Marihuana and Tobacco Smokers. Clin. Pharmacol. Ther. 1978, 24, 405–410. [Google Scholar] [CrossRef]
  69. Sands, T.T.; Rahdari, S.; Oldham, M.S.; Caminha Nunes, E.; Tilton, N.; Cilio, M.R. Long-Term Safety, Tolerability, and Efficacy of Cannabidiol in Children with Refractory Epilepsy: Results from an Expanded Access Program in the US. CNS Drugs 2019, 33, 47–60. [Google Scholar] [CrossRef]
  70. 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. [Google Scholar] [CrossRef]
  71. Brown, S.A.; Boyd, K.; Thomason, A.R. Geriatric Pharmacotherapy Case Series: Potential Drug Interactions with Antiplatelet Medications and Cannabidiol-A Focus on P2Y12 Inhibitors. Sr. Care Pharm. 2023, 38, 141–147. [Google Scholar] [CrossRef]
  72. Smythe, M.A.; Wu, W.; Garwood, C.L. Anticoagulant Drug–Drug Interactions with Cannabinoids: A Systematic Review. Pharmacotherapy 2023, 43, 1327–1338. [Google Scholar] [CrossRef]
  73. Kosel, B.W.; Aweeka, F.T.; Benowitz, N.L.; Shade, S.B.; Hilton, J.F.; Lizak, P.S.; Abrams, D.I. The Effects of Cannabinoids on the Pharmacokinetics of Indinavir and Nelfinavir. AIDS 2002, 16, 543–550. [Google Scholar] [CrossRef] [PubMed]
  74. Madden, K.; Tanco, K.; Bruera, E. Clinically Significant Drug-Drug Interaction between Methadone and Cannabidiol. Pediatrics 2020, 145, e20193256. [Google Scholar] [CrossRef]
  75. Manini, A.F.; Yiannoulos, G.; Bergamaschi, M.M.; Hernandez, S.; Olmedo, R.; Barnes, A.J.; Winkel, G.; Sinha, R.; Jutras-Aswad, D.; Huestis, M.A.; et al. Safety and Pharmacokinetics of Oral Cannabidiol When Administered Concomitantly with Intravenous Fentanyl in Humans. J. Addict. Med. 2015, 9, 204–210. [Google Scholar] [CrossRef] [PubMed]
  76. Roberts, J.D.; Gennings, C.; Shih, M. Synergistic Affective Analgesic Interaction between Delta-9-Tetrahydrocannabinol and Morphine. Eur. J. Pharmacol. 2006, 530, 54–58. [Google Scholar] [CrossRef]
  77. Murray, C.H.; Haney, M.; Foltin, R.W.; Manubay, J.; Bedi, G.; Cooper, Z.D. Smoked Cannabis Reduces Peak Cocaine Plasma Levels and Subjective Effects in a Controlled Drug Administration Study of Polysubstance Use in Men. Drug Alcohol. Depend. 2023, 243, 109757. [Google Scholar] [CrossRef]
  78. Beers, J.L.; Fu, D.; Jackson, K.D. Cytochrome P450-Catalyzed Metabolism of Cannabidiol to the Active Metabolite 7-Hydroxy-Cannabidiol. Drug Metab. Dispos. 2021, 49, 882–891. [Google Scholar] [CrossRef] [PubMed]
  79. Beers, J.L.; Authement, A.K.; Isoherranen, N.; Jackson, K.D. Cytosolic Enzymes Generate Cannabinoid Metabolites 7-Carboxycannabidiol and 11-Nor-9-Carboxytetrahydrocannabinol. ACS Med. Chem. Lett. 2023, 14, 614–620. [Google Scholar] [CrossRef]
  80. Auzmendi, J.; Palestro, P.; Blachman, A.; Gavernet, L.; Merelli, A.; Talevi, A.; Calabrese, G.C.; Ramos, A.J.; Lazarowski, A. Cannabidiol (CBD) Inhibited Rhodamine-123 Efflux in Cultured Vascular Endothelial Cells and Astrocytes Under Hypoxic Conditions. Front. Behav. Neurosci. 2020, 14, 32. [Google Scholar] [CrossRef]
  81. Brzozowska, N.; Li, K.M.; Wang, X.S.; Booth, J.; Stuart, J.; McGregor, I.S.; Arnold, J.C. ABC Transporters P-Gp and Bcrp Do Not Limit the Brain Uptake of the Novel Antipsychotic and Anticonvulsant Drug Cannabidiol in Mice. PeerJ 2016, 4, e2081. [Google Scholar] [CrossRef]
  82. Holland, M.L.; Allen, J.D.; Arnold, J.C. Interaction of Plant Cannabinoids with the Multidrug Transporter ABCC1 (MRP1). Eur. J. Pharmacol. 2008, 591, 128–131. [Google Scholar] [CrossRef]
  83. 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, 1415–1419. [Google Scholar] [CrossRef]
  84. Kumar, A.R.; Patilea-Vrana, G.I.; Anoshchenko, O.; Unadkat, J.D. Characterizing and Quantifying Extrahepatic Metabolism of (−)-Δ9-Tetrahydrocannabinol (THC) and Its Psychoactive Metabolite, (±)-11-Hydroxy-Δ9-THC (11-OH-THC). Drug Metab. Dispos. 2022, 50, 734–740. [Google Scholar] [CrossRef] [PubMed]
  85. Nasrin, S.; Watson, C.J.W.; Perez-Paramo, Y.X.; Lazarus, P. Cannabinoid Metabolites as Inhibitors of Major Hepatic CYP450 Enzymes, with Implications for Cannabis-Drug Interactions. Drug Metab. Dispos. 2021, 49, 1070–1080. [Google Scholar] [CrossRef]
  86. Center for Drug Evaluation and Research (CDER). Clinical Pharmacology and Biopharmaceutics Review (Cannabidiol) [Internet]. U.S. Food and Drug Administration Drugs@FDA. 2017. Available online: https://www.Accessdata.Fda.Gov/Drugsatfda_docs/Nda/2018/210365Orig1s000ClinPharmR.Pdf (accessed on 4 January 2024).
  87. Nasrin, S.; Watson, C.J.W.; Bardhi, K.; Fort, G.; Chen, G.; Lazarus, P. Inhibition of UDP-Glucuronosyltransferase Enzymes by Major Cannabinoids and Their Metabolites. Drug Metab. Dispos. 2021, 49, 1081–1089. [Google Scholar] [CrossRef]
  88. Alsherbiny, M.; Li, C. Medicinal Cannabis—Potential Drug Interactions. Medicines 2018, 6, 3. [Google Scholar] [CrossRef]
  89. Bland, T.M.; Haining, R.L.; Tracy, T.S.; Callery, P.S. CYP2C-Catalyzed Delta9-Tetrahydrocannabinol Metabolism: Kinetics, Pharmacogenetics and Interaction with Phenytoin. Biochem. Pharmacol. 2005, 70, 1096–1103. [Google Scholar] [CrossRef] [PubMed]
  90. Bonhomme-Faivre, L.; Benyamina, A.; Reynaud, M.; Farinotti, R.; Abbara, C. PRECLINICAL STUDY: Disposition of Δ9Tetrahydrocannabinol in CF1 Mice Deficient in Mdr1a P-glycoprotein. Addict. Biol. 2008, 13, 295–300. [Google Scholar] [CrossRef] [PubMed]
  91. Spiro, A.S.; Wong, A.; Boucher, A.A.; Arnold, J.C. Enhanced Brain Disposition and Effects of Δ9-Tetrahydrocannabinol in P-Glycoprotein and Breast Cancer Resistance Protein Knockout Mice. PLoS ONE 2012, 7, e35937. [Google Scholar] [CrossRef] [PubMed]
  92. Chen, X.; Unadkat, J.D.; Mao, Q. Tetrahydrocannabinol and Its Major Metabolites Are Not (or Are Poor) Substrates or Inhibitors of Human P-Glycoprotein [ATP-Binding Cassette (ABC) B1] and Breast Cancer Resistance Protein (ABCG2). Drug Metab. Dispos. 2021, 49, 910–918. [Google Scholar] [CrossRef] [PubMed]
  93. Chen, X.; Unadkat, J.D.; Mao, Q. Maternal and Fetal Exposure to (-)-Δ9-Tetrahydrocannabinol and Its Major Metabolites in Pregnant Mice Is Differentially Impacted by P-Glycoprotein and Breast Cancer Resistance Protein. Drug Metab. Dispos. 2023, 51, 269–275. [Google Scholar] [CrossRef]
  94. Qian, Y.; Markowitz, J.S. Prediction of Carboxylesterase 1-Mediated In Vivo Drug Interaction between Methylphenidate and Cannabinoids Using Static and Physiologically Based Pharmacokinetic Models. Drug Metab. Dispos. 2022, 50, 968–979. [Google Scholar] [CrossRef] [PubMed]
  95. Brzozowska, N.I.; de Tonnerre, E.J.; Li, K.M.; Wang, X.S.; Boucher, A.A.; Callaghan, P.D.; Kuligowski, M.; Wong, A.; Arnold, J.C. The Differential Binding of Antipsychotic Drugs to the ABC Transporter P-Glycoprotein Predicts Cannabinoid-Antipsychotic Drug Interactions. Neuropsychopharmacology 2017, 42, 2222–2231. [Google Scholar] [CrossRef] [PubMed]
  96. Giraud, C.; Tran, A.; Rey, E.; Vincent, J.; Tréluyer, J.-M.; Pons, G. In Vitro Characterization of Clobazam Metabolism by Recombinant Cytochrome P450 Enzymes: Importance of CYP2C19. Drug Metab. Dispos. 2004, 32, 1279–1286. [Google Scholar] [CrossRef]
  97. Thomas, T.F.; Metaxas, E.S.; Nguyen, T.; Bennett, W.; Skiendzielewski, K.V.; Quinn, D.H.; Scaletta, A.L. Case Report: Medical Cannabis-Warfarin Drug-Drug Interaction. J. Cannabis Res. 2022, 4, 6. [Google Scholar] [CrossRef] [PubMed]
  98. Brown, G.W.; Bellnier, T.J.; Janda, M.; Miskowitz, K. Δ-9-Tetrahydrocannabinol Dose Increase Leads to Warfarin Drug Interaction and Elevated INR. J. Am. Pharm. Assoc. 2021, 61, e57–e60. [Google Scholar] [CrossRef] [PubMed]
  99. Zamarripa, C.A.; Spindle, T.R.; Surujunarain, R.; Weerts, E.M.; Bansal, S.; Unadkat, J.D.; Paine, M.F.; Vandrey, R. Assessment of Orally Administered Δ9-Tetrahydrocannabinol When Coadministered with Cannabidiol on Δ9-Tetrahydrocannabinol Pharmacokinetics and Pharmacodynamics in Healthy Adults: A Randomized Clinical Trial. JAMA Netw. Open 2023, 6, e2254752. [Google Scholar] [CrossRef] [PubMed]
  100. FDA Center for Drug Evaluation and Research. Clinical Interaction Studies—Cytochrome P450 Enzyme- and Transporter-Mediated Drug Interactions: Guidance for Industry; Food and Drug Administration: Rockville, MD, USA, 2020.
  101. Engels, F.K.; de Jong, F.A.; Sparreboom, A.; Mathot, R.A.A.; Loos, W.J.; Kitzen, J.J.E.M.; de Bruijn, P.; Verweij, J.; Mathijssen, R.H.J. Medicinal Cannabis Does Not Influence the Clinical Pharmacokinetics of Irinotecan and Docetaxel. Oncologist 2007, 12, 291–300. [Google Scholar] [CrossRef]
  102. Riggs, C.E.; Egorin, M.J.; Fuks, J.Z.; Schnaper, N.; Duffey, P.; Colvin, O.M.; Aisner, J.; Wiernik, P.H.; Bachur, N.R. Initial Observations on the Effects of Δ9-Tetrahydrocannabinol on the Plasma Pharmacokinetics of Cyclophosphamide and Doxorubicin. J. Clin. Pharmacol. 1981, 21, 90S–98S. [Google Scholar] [CrossRef] [PubMed]
  103. Markowitz, J.S.; De Faria, L.; Zhang, Q.; Melchert, P.W.; Frye, R.F.; Klee, B.O.; Qian, Y. The Influence of Cannabidiol on the Pharmacokinetics of Methylphenidate in Healthy Subjects. Med. Cannabis Cannabinoids 2022, 5, 199–206. [Google Scholar] [CrossRef] [PubMed]
  104. Corso, B.; Bacle, A.; Demay, E.; Mercerolle, M.; Pelletier, R.; Gicquel, T.; Le Daré, B. Place of Therapeutic Cannabis in France and Safety Data: A Literature Review. Ann. Pharm. Fr. 2023, 81, 583–595. [Google Scholar] [CrossRef] [PubMed]
  105. Garrett, E.R.; Hunt, C.A. Physiochemical Properties, Solubility, and Protein Binding of Δ9-Tetrahydrocannabinol. J. Pharm. Sci. 1974, 63, 1056–1064. [Google Scholar] [CrossRef] [PubMed]
  106. Golombek, P.; Müller, M.; Barthlott, I.; Sproll, C.; Lachenmeier, D.W. Conversion of Cannabidiol (CBD) into Psychotropic Cannabinoids Including Tetrahydrocannabinol (THC): A Controversy in the Scientific Literature. Toxics 2020, 8, 41. [Google Scholar] [CrossRef]
  107. Merrick, J.; Lane, B.; Sebree, T.; Yaksh, T.; O’Neill, C.; Banks, S.L. Identification of Psychoactive Degradants of Cannabidiol in Simulated Gastric and Physiological Fluid. Cannabis Cannabinoid Res. 2016, 1, 102–112. [Google Scholar] [CrossRef]
  108. Hložek, T.; Uttl, L.; Kadeřábek, L.; Balíková, M.; Lhotková, E.; Horsley, R.R.; Nováková, P.; Šíchová, K.; Štefková, K.; Tylš, F.; et al. Pharmacokinetic and Behavioural Profile of THC, CBD, and THC+CBD Combination after Pulmonary, Oral, and Subcutaneous Administration in Rats and Confirmation of Conversion in Vivo of CBD to THC. Eur. Neuropsychopharmacol. 2017, 27, 1223–1237. [Google Scholar] [CrossRef] [PubMed]
  109. Crippa, J.A.S.; Zuardi, A.W.; Hallak, J.E.C.; Miyazawa, B.; Bernardo, S.A.; Donaduzzi, C.M.; Guzzi, S.; Favreto, W.A.J.; Campos, A.; Queiroz, M.E.C.; et al. Oral Cannabidiol Does Not Convert to Δ8-THC or Δ9-THC in Humans: A Pharmacokinetic Study in Healthy Subjects. Cannabis Cannabinoid Res. 2020, 5, 89–98. [Google Scholar] [CrossRef] [PubMed]
  110. Bruckmueller, H.; Cascorbi, I. Drug-Drug-Gene Interactions: A Call for Clinical Consideration. Clin. Pharma Ther. 2021, 110, 549–551. [Google Scholar] [CrossRef]
  111. Bahar, M.A.; Setiawan, D.; Hak, E.; Wilffert, B. Pharmacogenetics of Drug–Drug Interaction and Drug–Drug–Gene Interaction: A Systematic Review on CYP2C9, CYP2C19 and CYP2D6. Pharmacogenomics 2017, 18, 701–739. [Google Scholar] [CrossRef]
  112. Zanger, U.M.; Schwab, M. Cytochrome P450 Enzymes in Drug Metabolism: Regulation of Gene Expression, Enzyme Activities, and Impact of Genetic Variation. Pharmacol. Ther. 2013, 138, 103–141. [Google Scholar] [CrossRef]
  113. Zhou, S.-F.; Liu, J.-P.; Chowbay, B. Polymorphism of Human Cytochrome P450 Enzymes and Its Clinical Impact. Drug Metab. Rev. 2009, 41, 89–295. [Google Scholar] [CrossRef]
  114. Wolowich, W.R.; Greif, R.; Kleine-Brueggeney, M.; Bernhard, W.; Theiler, L. Minimal Physiologically Based Pharmacokinetic Model of Intravenously and Orally Administered Delta-9-Tetrahydrocannabinol in Healthy Volunteers. Eur. J. Drug Metab. Pharmacokinet. 2019, 44, 691–711. [Google Scholar] [CrossRef]
  115. Cáceres Guido, P.; Riva, N.; Caraballo, R.; Reyes, G.; Huaman, M.; Gutierrez, R.; Agostini, S.; Fabiana Delaven, S.; Pérez Montilla, C.A.; García Bournissen, F.; et al. Pharmacokinetics of Cannabidiol in Children with Refractory Epileptic Encephalopathy. Epilepsia 2021, 62, e7–e12. [Google Scholar] [CrossRef]
  116. Wang, G.S.; Bourne, D.W.A.; Klawitter, J.; Sempio, C.; Chapman, K.; Knupp, K.; Wempe, M.F.; Borgelt, L.; Christians, U.; Leonard, J.; et al. Disposition of Oral Cannabidiol-Rich Cannabis Extracts in Children with Epilepsy. Clin. Pharmacokinet. 2020, 59, 1005–1012. [Google Scholar] [CrossRef]
  117. 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, 1110–1119. [Google Scholar] [CrossRef]
  118. Tayo, B.; Taylor, L.; Sahebkar, F.; Morrison, G. A Phase I, 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. 2020, 59, 747–755. [Google Scholar] [CrossRef]
  119. Clarke, S.; Butcher, B.E.; McLachlan, A.J.; Henson, J.D.; Rutolo, D.; Hall, S.; Vitetta, L. Pilot Clinical and Pharmacokinetic Study of Δ9-Tetrahydrocannabinol (THC)/Cannabidiol (CBD) Nanoparticle Oro-Buccal Spray in Patients with Advanced Cancer Experiencing Uncontrolled Pain. PLoS ONE 2022, 17, e0270543. [Google Scholar] [CrossRef]
  120. Birnbaum, A.K.; Karanam, A.; Marino, S.E.; Barkley, C.M.; Remmel, R.P.; Roslawski, M.; Gramling-Aden, M.; Leppik, I.E. Food Effect on Pharmacokinetics of Cannabidiol Oral Capsules in Adult Patients with Refractory Epilepsy. Epilepsia 2019, 60, 1586–1592. [Google Scholar] [CrossRef]
  121. Hansen, J.S.; Boix, F.; Hasselstrøm, J.B.; Sørensen, L.K.; Kjolby, M.; Gustavsen, S.; Hansen, R.M.; Petersen, T.; Sellebjerg, F.; Kasch, H.; et al. Pharmacokinetics and Pharmacodynamics of Cannabis-based Medicine in a Patient Population Included in a Randomized, Placebo-controlled, Clinical Trial. Clin. Transl. Sci. 2024, 17, e13685. [Google Scholar] [CrossRef]
  122. Naftali, T.; Bar-Lev Schleider, L.; Almog, S.; Meiri, D.; Konikoff, F.M. Cannabis Is Associated with Clinical but Not Endoscopic Remission in Ulcerative Colitis: A Randomized Controlled Trial. J. Crohns Colitis 2021, 15, 1799–1806. [Google Scholar] [CrossRef]
  123. Ahmed, A.I.A.; van den Elsen, G.A.H.; Colbers, A.; van der Marck, M.A.; Burger, D.M.; Feuth, T.B.; Rikkert, M.G.M.O.; Kramers, C. Safety and Pharmacokinetics of Oral Delta-9-Tetrahydrocannabinol in Healthy Older Subjects: A Randomized Controlled Trial. Eur. Neuropsychopharmacol. 2014, 24, 1475–1482. [Google Scholar] [CrossRef]
  124. Ahmed, A.I.A.; van den Elsen, G.A.H.; Colbers, A.; Kramers, C.; Burger, D.M.; van der Marck, M.A.; Olde Rikkert, M.G.M. Safety, Pharmacodynamics, and Pharmacokinetics of Multiple Oral Doses of Delta-9-Tetrahydrocannabinol in Older Persons with Dementia. Psychopharmacology 2015, 232, 2587–2595. [Google Scholar] [CrossRef]
  125. Almog, S.; Aharon-Peretz, J.; Vulfsons, S.; Ogintz, M.; Abalia, H.; Lupo, T.; Hayon, Y.; Eisenberg, E. The Pharmacokinetics, Efficacy, and Safety of a Novel Selective-Dose Cannabis Inhaler in Patients with Chronic Pain: A Randomized, Double-Blinded, Placebo-Controlled Trial. Eur. J. Pain 2020, 24, 1505–1516. [Google Scholar] [CrossRef]
  126. Eisenberg, E.; Ogintz, M.; Almog, S. The Pharmacokinetics, Efficacy, Safety, and Ease of Use of a Novel Portable Metered-Dose Cannabis Inhaler in Patients with Chronic Neuropathic Pain: A Phase 1a Study. J. Pain. Palliat. Care Pharmacother. 2014, 28, 216–225. [Google Scholar] [CrossRef]
  127. Volkow, N.D.; Han, B.; Compton, W.M.; McCance-Katz, E.F. Self-Reported Medical and Nonmedical Cannabis Use among Pregnant Women in the United States. JAMA 2019, 322, 167–169. [Google Scholar] [CrossRef]
  128. Young-Wolff, K.C.; Tucker, L.-Y.; Alexeeff, S.; Armstrong, M.A.; Conway, A.; Weisner, C.; Goler, N. Trends in Self-Reported and Biochemically Tested Marijuana Use among Pregnant Females in California from 2009–2016. JAMA 2017, 318, 2490–2491. [Google Scholar] [CrossRef]
  129. Monfort, A.; Ferreira, E.; Leclair, G.; Lodygensky, G.A. Pharmacokinetics of Cannabis and Its Derivatives in Animals and Humans During Pregnancy and Breastfeeding. Front. Pharmacol. 2022, 13, 919630. [Google Scholar] [CrossRef]
  130. U.S. Food and Drug Administration. Safety of CBD in Humans—A Literature Review (As of 12 December 2019). Available online: https://www.fda.gov/media/152317/download (accessed on 24 January 2024).
  131. Metz, T.D.; McMillin, G.A.; Silver, R.M.; Allshouse, A.A.; Heard, K.; Jensen, T.L.; Wymore, E.M.; Stickrath, E.; Conageski, C.; Kinney, G.L.; et al. Quantification of Prenatal Marijuana Use: Evaluation of the Correlation between Self-Report, Serum, Urine and Umbilical Cord Assays among Women Delivering at Two Urban Colorado Hospitals. Addiction 2022, 117, 172–181. [Google Scholar] [CrossRef]
  132. Westin, A.A.; Huestis, M.A.; Aarstad, K.; Spigset, O. Short Communication: Urinary Excretion of 11-nor-9-Carboxy-Δ9-Tetrahydrocannabinol in a Pregnant Woman Following Heavy, Chronic Cannabis Use. J. Anal. Toxicol. 2009, 33, 610–614. [Google Scholar] [CrossRef]
  133. Baker, T.; Datta, P.; Rewers-Felkins, K.; Thompson, H.; Kallem, R.R.; Hale, T.W. Transfer of Inhaled Cannabis Into Human Breast Milk. Obs. Gynecol. 2018, 131, 783–788. [Google Scholar] [CrossRef]
  134. Bertrand, K.A.; Hanan, N.J.; Honerkamp-Smith, G.; Best, B.M.; Chambers, C.D. Marijuana Use by Breastfeeding Mothers and Cannabinoid Concentrations in Breast Milk. Pediatrics 2018, 142, e20181076. [Google Scholar] [CrossRef]
  135. Sujan, A.C.; Pal, A.; Avalos, L.A.; Young-Wolff, K.C. A Systematic Review of in Utero Cannabis Exposure and Risk for Structural Birth Defects. Front. Pediatr. 2023, 11, 1149401. [Google Scholar] [CrossRef]
  136. Dochez-Arnault, J.; Desdoits-Lethimonier, C.; Matias, I.; Evrard, B.; Lagarrigue, M.; Toupin, M.; Lardenois, A.; Chalmel, F.; Mazaud-Guittot, S.; Dejucq-Rainsford, N.; et al. Expression of the Endocannabinoid System and Response to Cannabinoid Components by the Human Fetal Testis. BMC Med. 2023, 21, 219. [Google Scholar] [CrossRef]
  137. Patilea-Vrana, G.I.; Unadkat, J.D. Development and Verification of a Linked Δ9-THC/11-OH-THC Physiologically Based Pharmacokinetic Model in Healthy, Nonpregnant Population and Extrapolation to Pregnant Women. Drug Metab. Dispos. 2021, 49, 509–520. [Google Scholar] [CrossRef]
  138. Shenkoya, B.; Yellepeddi, V.; Mark, K.; Gopalakrishnan, M. Predicting Maternal and Infant Tetrahydrocannabinol Exposure in Lactating Cannabis Users: A Physiologically Based Pharmacokinetic Modeling Approach. Pharmaceutics 2023, 15, 2467. [Google Scholar] [CrossRef]
  139. Trivett, C.; Lees, Z.J.; Freeman, D.J. Adipose Tissue Function in Healthy Pregnancy, Gestational Diabetes Mellitus and Pre-Eclampsia. Eur. J. Clin. Nutr. 2021, 75, 1745–1756. [Google Scholar] [CrossRef]
  140. Pipe, N.G.; Smith, T.; Halliday, D.; Edmonds, C.J.; Williams, C.; Coltart, T.M. Changes in Fat, Fat-Free Mass and Body Water in Human Normal Pregnancy. Br. J. Obs. Gynaecol. 1979, 86, 929–940. [Google Scholar] [CrossRef]
  141. Grant, K.S.; Petroff, R.; Isoherranen, N.; Stella, N.; Burbacher, T.M. Cannabis Use during Pregnancy: Pharmacokinetics and Effects on Child Development. Pharmacol. Ther. 2018, 182, 133–151. [Google Scholar] [CrossRef]
  142. Carlier, J.; Huestis, M.A.; Zaami, S.; Pichini, S.; Busardò, F.P. Monitoring Perinatal Exposure to Cannabis and Synthetic Cannabinoids. Ther. Drug Monit. 2020, 42, 194–204. [Google Scholar] [CrossRef]
  143. Sorkhou, M.; Singla, D.R.; Castle, D.J.; George, T.P. Birth, Cognitive and Behavioral Effects of Intrauterine Cannabis Exposure in Infants and Children: A Systematic Review and Meta-Analysis. Addiction 2023, 119, 411–437. [Google Scholar] [CrossRef]
  144. Aran, A.; Cayam-Rand, D. Medical Cannabis in Children. Rambam Maimonides Med. J. 2020, 11, e0003. [Google Scholar] [CrossRef]
  145. Malach, M.; Kovalchuk, I.; Kovalchuk, O. Medical Cannabis in Pediatric Oncology: Friend or Foe? Pharmaceuticals 2022, 15, 359. [Google Scholar] [CrossRef]
  146. Hacohen, M.; Stolar, O.E.; Berkovitch, M.; Elkana, O.; Kohn, E.; Hazan, A.; Heyman, E.; Sobol, Y.; Waissengreen, D.; Gal, E.; et al. Children and Adolescents with ASD Treated with CBD-Rich Cannabis Exhibit Significant Improvements Particularly in Social Symptoms: An Open Label Study. Transl. Psychiatry 2022, 12, 375. [Google Scholar] [CrossRef]
  147. Efron, D.; Taylor, K.; Payne, J.M.; Freeman, J.L.; Cranswick, N.; Mulraney, M.; Prakash, C.; Lee, K.J.; Williams, K. Does Cannabidiol Reduce Severe Behavioural Problems in Children with Intellectual Disability? Study Protocol for a Pilot Single-Site Phase I/II Randomised Placebo Controlled Trial. BMJ Open 2020, 10, e034362. [Google Scholar] [CrossRef]
  148. Heussler, H.; Cohen, J.; Silove, N.; Tich, N.; Bonn-Miller, M.O.; Du, W.; O’Neill, C.; Sebree, T. A Phase 1/2, Open-Label Assessment of the Safety, Tolerability, and Efficacy of Transdermal Cannabidiol (ZYN002) for the Treatment of Pediatric Fragile X Syndrome. J. Neurodev. Disord. 2019, 11, 16. [Google Scholar] [CrossRef]
  149. Libzon, S.; Schleider, L.B.-L.; Saban, N.; Levit, L.; Tamari, Y.; Linder, I.; Lerman-Sagie, T.; Blumkin, L. Medical Cannabis for Pediatric Moderate to Severe Complex Motor Disorders. J. Child. Neurol. 2018, 33, 565–571. [Google Scholar] [CrossRef]
  150. Ekert, H.; Waters, K.D.; Jurk, I.H.; Mobilia, J.; Loughnan, P. Amelioration of Cancer Chemotherapy-Induced Nausea and Vomiting by Delta-9-Tetrahydrocannabinol. Med. J. Aust. 1979, 2, 657–659. [Google Scholar] [CrossRef]
  151. Fairhurst, C.; Kumar, R.; Checketts, D.; Tayo, B.; Turner, S. Efficacy and Safety of Nabiximols Cannabinoid Medicine for Paediatric Spasticity in Cerebral Palsy or Traumatic Brain Injury: A Randomized Controlled Trial. Dev. Med. Child. Neurol. 2020, 62, 1031–1039. [Google Scholar] [CrossRef]
  152. Treves, N.; Mor, N.; Allegaert, K.; Bassalov, H.; Berkovitch, M.; Stolar, O.E.; Matok, I. Efficacy and Safety of Medical Cannabinoids in Children: A Systematic Review and Meta-Analysis. Sci. Rep. 2021, 11, 23462. [Google Scholar] [CrossRef]
  153. Scheffer, I.E.; Halford, J.J.; Miller, I.; Nabbout, R.; Sanchez-Carpintero, R.; Shiloh-Malawsky, Y.; Wong, M.; Zolnowska, M.; Checketts, D.; Dunayevich, E.; et al. Add-on Cannabidiol in Patients with Dravet Syndrome: Results of a Long-term Open-label Extension Trial. Epilepsia 2021, 62, 2505–2517. [Google Scholar] [CrossRef]
  154. Smid, M.C.; Metz, T.D.; McMillin, G.A.; Mele, L.; Casey, B.M.; Reddy, U.M.; Wapner, R.J.; Thorp, J.M.; Saade, G.R.; Tita, A.T.N.; et al. Prenatal Nicotine or Cannabis Exposure and Offspring Neurobehavioral Outcomes. Obs. Gynecol. 2022, 139, 21–30. [Google Scholar] [CrossRef]
  155. Devinsky, O.; Patel, A.D.; Thiele, E.A.; Wong, M.H.; Appleton, R.; Harden, C.L.; Greenwood, S.; Morrison, G.; Sommerville, K. Randomized, Dose-Ranging Safety Trial of Cannabidiol in Dravet Syndrome. Neurology 2018, 90, e1204–e1211. [Google Scholar] [CrossRef] [PubMed]
  156. Devinsky, O.; Kraft, K.; Rusch, L.; Fein, M.; Leone-Bay, A. Improved Bioavailability with Dry Powder Cannabidiol Inhalation: A Phase 1 Clinical Study. J. Pharm. Sci. 2021, 110, 3946–3952. [Google Scholar] [CrossRef]
  157. 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, 267–277. [Google Scholar] [CrossRef] [PubMed]
  158. Bansal, S.; Ladumor, M.K.; Paine, M.F.; Unadkat, J.D. A Physiologically-Based Pharmacokinetic Model for Cannabidiol in Healthy Adults, Hepatically-Impaired Adults, and Children. Drug Metab. Dispos. 2023, 51, 743–752. [Google Scholar] [CrossRef]
  159. Brussee, J.M.; Yu, H.; Krekels, E.H.J.; De Roos, B.; Brill, M.J.E.; Van Den Anker, J.N.; Rostami-Hodjegan, A.; De Wildt, S.N.; Knibbe, C.A.J. First-Pass CYP3A-Mediated Metabolism of Midazolam in the Gut Wall and Liver in Preterm Neonates. CPT Pharmacomet. Syst. Pharmacol. 2018, 7, 374–383. [Google Scholar] [CrossRef]
  160. Morselli, P.L.; Franco-Morselli, R.; Bossi, L. Clinical Pharmacokinetics in Newborns and Infants Age-Related Differences and Therapeutic Implications. Clin. Pharmacokinet. 1980, 5, 485–527. [Google Scholar] [CrossRef] [PubMed]
  161. Fernandez, E.; Perez, R.; Hernandez, A.; Tejada, P.; Arteta, M.; Ramos, J.T. Factors and Mechanisms for Pharmacokinetic Differences between Pediatric Population and Adults. Pharmaceutics 2011, 3, 53–72. [Google Scholar] [CrossRef]
  162. Strolin Benedetti, M.; Whomsley, R.; Baltes, E.L. Differences in Absorption, Distribution, Metabolism and Excretion of Xenobiotics between the Paediatric and Adult Populations. Expert. Opin. Drug Metab. Toxicol. 2005, 1, 447–471. [Google Scholar] [CrossRef]
  163. Nicolas, J.-M.; de Lange, E.C.M. Mind the Gaps: Ontogeny of Human Brain P-Gp and Its Impact on Drug Toxicity. AAPS J. 2019, 21, 67. [Google Scholar] [CrossRef] [PubMed]
  164. Kuzawa, C.W. Adipose Tissue in Human Infancy and Childhood: An Evolutionary Perspective. Am. J. Phys. Anthropol. 1998, 107, 177–209. [Google Scholar] [CrossRef]
  165. Butte, N.F.; Hopkinson, J.M.; Wong, W.W.; Smith, E.O.; Ellis, K.J. Body Composition during the First 2 Years of Life: An Updated Reference. Pediatr. Res. 2000, 47, 578–585. [Google Scholar] [CrossRef]
  166. Maharaj, A.R.; Gonzalez, D.; Cohen-Wolkowiez, M.; Hornik, C.P.; Edginton, A.N. Improving Pediatric Protein Binding Estimates: An Evaluation of A1-Acid Glycoprotein Maturation in Healthy and Infected Subjects. Clin. Pharmacokinet. 2018, 57, 577–589. [Google Scholar] [CrossRef] [PubMed]
  167. Goasdoué, K.; Miller, S.M.; Colditz, P.B.; Björkman, S.T. Review: The Blood-Brain Barrier; Protecting the Developing Fetal Brain. Placenta 2017, 54, 111–116. [Google Scholar] [CrossRef] [PubMed]
  168. de Wildt, S.N.; Kearns, G.L.; Leeder, J.S.; van den Anker, J.N. Cytochrome P450 3A: Ontogeny and Drug Disposition. Clin. Pharmacokinet. 1999, 37, 485–505. [Google Scholar] [CrossRef] [PubMed]
  169. Koukouritaki, S.B.; Manro, J.R.; Marsh, S.A.; Stevens, J.C.; Rettie, A.E.; McCarver, D.G.; Hines, R.N. Developmental Expression of Human Hepatic CYP2C9 and CYP2C19. J. Pharmacol. Exp. Ther. 2004, 308, 965–974. [Google Scholar] [CrossRef]
  170. Hines, R.N. Ontogeny of Human Hepatic Cytochromes P450. J. Biochem. Mol. Toxicol. 2007, 21, 169–175. [Google Scholar] [CrossRef]
  171. Miura, H. Zonisamide Monotherapy with Once-Daily Dosing in Children with Cryptogenic Localization-Related Epilepsies: Clinical Effects and Pharmacokinetic Studies. Seizure 2004, 13 (Suppl. S1), S17–S23; discussion S24–S25. [Google Scholar] [CrossRef]
  172. Vaughn, S.E.; Strawn, J.R.; Poweleit, E.A.; Sarangdhar, M.; Ramsey, L.B. The Impact of Marijuana on Antidepressant Treatment in Adolescents: Clinical and Pharmacologic Considerations. J. Pers. Med. 2021, 11, 615. [Google Scholar] [CrossRef]
  173. Aftab, A.; Lam, J.A.; Liu, F.; Ghosh, A.; Sajatovic, M. Recent Developments in Geriatric Psychopharmacology. Expert. Rev. Clin. Pharmacol. 2021, 14, 341–355. [Google Scholar] [CrossRef]
  174. Fonseca, C.; Ettcheto, M.; Bicker, J.; Fernandes, M.J.; Falcão, A.; Camins, A.; Fortuna, A. Under the Umbrella of Depression and Alzheimer’s Disease Physiopathology: Can Cannabinoids Be a Dual-Pleiotropic Therapy? Ageing Res. Rev. 2023, 90, 101998. [Google Scholar] [CrossRef] [PubMed]
  175. Kanchi, P.K.; Dasmahapatra, A.K. Destabilization of the Alzheimer’s Amyloid-β Protofibrils by THC: A Molecular Dynamics Simulation Study. J. Mol. Graph. Model. 2021, 105, 107889. [Google Scholar] [CrossRef] [PubMed]
  176. Watson, K.K.; Bryan, A.D.; Thayer, R.E.; Ellingson, J.M.; Skrzynski, C.J.; Hutchison, K.E. Cannabis Use and Resting State Functional Connectivity in the Aging Brain. Front. Aging Neurosci. 2022, 14, 804890. [Google Scholar] [CrossRef] [PubMed]
  177. Zhang, X.-B.; Li, J.; Gu, J.; Zeng, Y.-Q. Roles of Cannabidiol in the Treatment and Prevention of Alzheimer’sDisease by Multi-Target Actions. Mini Rev. Med. Chem. 2022, 22, 43–51. [Google Scholar] [CrossRef] [PubMed]
  178. Bianchi, F.; Pautex, S.; Wampfler, J.; Curtin, F.; Daali, Y.; Desmeules, J.A.; Broers, B. Medical Cannabinoids for Painful Symptoms in Patients with Severe Dementia: A Randomized, Double-Blind Cross-over Placebo-Controlled Trial Protocol. Front. Pain Res. 2023, 4, 1108832. [Google Scholar] [CrossRef] [PubMed]
  179. Pautex, S.; Bianchi, F.; Daali, Y.; Augsburger, M.; de Saussure, C.; Wampfler, J.; Curtin, F.; Desmeules, J.; Broers, B. Cannabinoids for Behavioral Symptoms in Severe Dementia: Safety and Feasibility in a Long-Term Pilot Observational Study in Nineteen Patients. Front. Aging Neurosci. 2022, 14, 957665. [Google Scholar] [CrossRef] [PubMed]
  180. Costa, A.C.; Joaquim, H.P.G.; Pedrazzi, J.F.C.; Pain, A.D.O.; Duque, G.; Aprahamian, I. Cannabinoids in Late Life Parkinson’s Disease and Dementia: Biological Pathways and Clinical Challenges. Brain Sci. 2022, 12, 1596. [Google Scholar] [CrossRef] [PubMed]
  181. da Cruz Guedes, E.; Erustes, A.G.; Leão, A.H.F.F.; Carneiro, C.A.; Abílio, V.C.; Zuardi, A.W.; Hallak, J.E.C.; Crippa, J.A.; Bincoletto, C.; Smaili, S.S.; et al. Cannabidiol Recovers Dopaminergic Neuronal Damage Induced by Reserpine or α-Synuclein in Caenorhabditis Elegans. Neurochem. Res. 2023, 48, 2390–2405. [Google Scholar] [CrossRef]
  182. Hasumi, A.; Maeda, H. Cannabidiol Improves Haloperidol-Induced Motor Dysfunction in Zebrafish: A Comparative Study with a Dopamine Activating Drug. J. Cannabis Res. 2023, 5, 6. [Google Scholar] [CrossRef]
  183. Kluger, B.M.; Huang, A.P.; Miyasaki, J.M. Cannabinoids in Movement Disorders. Park. Relat. Disord. 2022, 102, 124–130. [Google Scholar] [CrossRef]
  184. Muhammad, F.; Liu, Y.; Wang, N.; Zhao, L.; Zhou, Y.; Yang, H.; Li, H. Neuroprotective Effects of Cannabidiol on Dopaminergic Neurodegeneration and α-Synuclein Accumulation in C. Elegans Models of Parkinson’s Disease. NeuroToxicology 2022, 93, 128–139. [Google Scholar] [CrossRef] [PubMed]
  185. Sadaka, A.H.; Canuel, J.; Febo, M.; Johnson, C.T.; Bradshaw, H.B.; Ortiz, R.; Ciumo, F.; Kulkarni, P.; Gitcho, M.A.; Ferris, C.F. Effects of Inhaled Cannabis High in Δ9-THC or CBD on the Aging Brain: A Translational MRI and Behavioral Study. Front. Aging Neurosci. 2023, 15, 1055433. [Google Scholar] [CrossRef] [PubMed]
  186. Twelves, C.; Sabel, M.; Checketts, D.; Miller, S.; Tayo, B.; Jove, M.; Brazil, L.; Short, S.C.; on behalf of the GWCA1208 Study Group. A Phase 1b Randomised, Placebo-Controlled Trial of Nabiximols Cannabinoid Oromucosal Spray with Temozolomide in Patients with Recurrent Glioblastoma. Br. J. Cancer 2021, 124, 1379–1387. [Google Scholar] [CrossRef] [PubMed]
  187. Frytak, S. Delta-9-Tetrahydrocannabinol as an Antiemetic for Patients Receiving Cancer Chemotherapy: A Comparison with Prochlorperazine and a Placebo. Ann. Intern. Med. 1979, 91, 825. [Google Scholar] [CrossRef] [PubMed]
  188. Dumic, I.; Nordin, T.; Jecmenica, M.; Stojkovic Lalosevic, M.; Milosavljevic, T.; Milovanovic, T. Gastrointestinal Tract Disorders in Older Age. Can. J. Gastroenterol. Hepatol. 2019, 2019, 6757524. [Google Scholar] [CrossRef] [PubMed]
  189. Stillhart, C.; Asteriadis, A.; Bocharova, E.; Eksteen, G.; Harder, F.; Kusch, J.; Tzakri, T.; Augustijns, P.; Matthys, C.; Vertzoni, M.; et al. The Impact of Advanced Age on Gastrointestinal Characteristics That Are Relevant to Oral Drug Absorption: An AGePOP Review. Eur. J. Pharm. Sci. 2023, 187, 106452. [Google Scholar] [CrossRef]
  190. Giezenaar, C.; Trahair, L.G.; Rigda, R.; Hutchison, A.T.; Feinle-Bisset, C.; Luscombe-Marsh, N.D.; Hausken, T.; Jones, K.L.; Horowitz, M.; Chapman, I.; et al. Lesser Suppression of Energy Intake by Orally Ingested Whey Protein in Healthy Older Men Compared with Young Controls. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2015, 309, R845–R854. [Google Scholar] [CrossRef] [PubMed]
  191. Pentafragka, C.; Vertzoni, M.; Symillides, M.; Goumas, K.; Reppas, C. Disposition of Two Highly Permeable Drugs in the Upper Gastrointestinal Lumen of Healthy Adults after a Standard High-Calorie, High-Fat Meal. Eur. J. Pharm. Sci. 2020, 149, 105351. [Google Scholar] [CrossRef]
  192. Evans, M.A.; Triggs, E.J.; Cheung, M.; Broe, G.A.; Creasey, H. Gastric Emptying Rate in the Elderly: Implications for Drug Therapy. J. Am. Geriatr. Soc. 1981, 29, 201–205. [Google Scholar] [CrossRef]
  193. Soenen, S.; Rayner, C.K.; Horowitz, M.; Jones, K.L. Gastric Emptying in the Elderly. Clin. Geriatr. Med. 2015, 31, 339–353. [Google Scholar] [CrossRef]
  194. Kataoka, M.; Fukahori, M.; Ikemura, A.; Kubota, A.; Higashino, H.; Sakuma, S.; Yamashita, S. Effects of Gastric pH on Oral Drug Absorption: In Vitro Assessment Using a Dissolution/Permeation System Reflecting the Gastric Dissolution Process. Eur. J. Pharm. Biopharm. 2016, 101, 103–111. [Google Scholar] [CrossRef] [PubMed]
  195. Manchikanti, L.; Colliver, J.A.; Marrero, T.C.; Roush, J.R. Assessment of Age-Related Acid Aspiration Risk Factors in Pediatric, Adult, and Geriatric Patients. Anesth. Analg. 1985, 64, 11–17. [Google Scholar] [CrossRef] [PubMed]
  196. Vertzoni, M.; Sulaiman, S.; Goumas, K.; Kersten, E.; Anlahr, J.; Muenster, U.; Reppas, C. Characteristics of Contents of Lower Intestine in the 65–74 Years of Age Range Could Impact the Performance of Safe and Efficacious Modified Release Products. J. Pharm. Sci. 2021, 110, 251–258. [Google Scholar] [CrossRef] [PubMed]
  197. Mahmood, A.; Bernkop-Schnürch, A. SEDDS: A Game Changing Approach for the Oral Administration of Hydrophilic Macromolecular Drugs. Adv. Drug Deliv. Rev. 2019, 142, 91–101. [Google Scholar] [CrossRef] [PubMed]
  198. Laugier, R.; Bernard, J.P.; Berthezene, P.; Dupuy, P. Changes in Pancreatic Exocrine Secretion with Age: Pancreatic Exocrine Secretion Does Decrease in the Elderly. Digestion 1991, 50, 202–211. [Google Scholar] [CrossRef] [PubMed]
  199. Cho, S.J.; Stout-Delgado, H.W. Aging and Lung Disease. Annu. Rev. Physiol. 2020, 82, 433–459. [Google Scholar] [CrossRef] [PubMed]
  200. Schneider, J.L.; Rowe, J.H.; Garcia-de-Alba, C.; Kim, C.F.; Sharpe, A.H.; Haigis, M.C. The Aging Lung: Physiology, Disease, and Immunity. Cell 2021, 184, 1990–2019. [Google Scholar] [CrossRef]
  201. Wallin, M.; Tagami, T.; Chen, L.; Yang, M.; Chan, H.-K. Pulmonary Drug Delivery to Older People. Adv. Drug Deliv. Rev. 2018, 135, 50–61. [Google Scholar] [CrossRef]
  202. Nagler, R.M.; Hershkovich, O. Age-Related Changes in Unstimulated Salivary Function and Composition and Its Relations to Medications and Oral Sensorial Complaints. Aging Clin. Exp. Res. 2005, 17, 358–366. [Google Scholar] [CrossRef]
  203. Nagler, R.M.; Hershkovich, O. Relationships between Age, Drugs, Oral Sensorial Complaints and Salivary Profile. Arch. Oral. Biol. 2005, 50, 7–16. [Google Scholar] [CrossRef]
  204. Ponti, F.; Santoro, A.; Mercatelli, D.; Gasperini, C.; Conte, M.; Martucci, M.; Sangiorgi, L.; Franceschi, C.; Bazzocchi, A. Aging and Imaging Assessment of Body Composition: From Fat to Facts. Front. Endocrinol. 2019, 10, 861. [Google Scholar] [CrossRef] [PubMed]
  205. Singam, N.S.V.; Fine, C.; Fleg, J.L. Cardiac Changes Associated with Vascular Aging. Clin. Cardiol. 2020, 43, 92–98. [Google Scholar] [CrossRef] [PubMed]
  206. Mangoni, A.A.; Jarmuzewska, E.A. The Influence of Heart Failure on the Pharmacokinetics of Cardiovascular and Non-cardiovascular Drugs: A Critical Appraisal of the Evidence. Br. J. Clin. Pharmacol. 2019, 85, 20–36. [Google Scholar] [CrossRef] [PubMed]
  207. Farrall, A.J.; Wardlaw, J.M. Blood–Brain Barrier: Ageing and Microvascular Disease—Systematic Review and Meta-Analysis. Neurobiol. Aging 2009, 30, 337–352. [Google Scholar] [CrossRef] [PubMed]
  208. Brock, F.; Bettinelli, L.A.; Dobner, T.; Stobbe, J.C.; Pomatti, G.; Telles, C.T. Prevalence of Hypoalbuminemia and Nutritional Issues in Hospitalized Elders. Rev. Lat.-Am. Enferm. 2016, 24, e2736. [Google Scholar] [CrossRef] [PubMed]
  209. Veering, B.T.; Burm, A.G.; Souverijn, J.H.; Serree, J.M.; Spierdijk, J. The Effect of Age on Serum Concentrations of Albumin and Alpha 1-Acid Glycoprotein. Br. J. Clin. Pharmacol. 1990, 29, 201–206. [Google Scholar] [CrossRef]
  210. Schmucker, D.L. Age-Related Changes in Liver Structure and Function: Implications for Disease ? Exp. Gerontol. 2005, 40, 650–659. [Google Scholar] [CrossRef]
  211. Wynne, H.A.; Cope, L.H.; Mutch, E.; Rawlins, M.D.; Woodhouse, K.W.; James, O.F. The Effect of Age upon Liver Volume and Apparent Liver Blood Flow in Healthy Man. Hepatology 1989, 9, 297–301. [Google Scholar] [CrossRef] [PubMed]
  212. Sotaniemi, E.A.; Arranto, A.J.; Pelkonen, O.; Pasanen, M. Age and Cytochrome P450-Linked Drug Metabolism in Humans: An Analysis of 226 Subjects with Equal Histopathologic Conditions. Clin. Pharmacol. Ther. 1997, 61, 331–339. [Google Scholar] [CrossRef]
  213. Kawase, A.; Ito, A.; Yamada, A.; Iwaki, M. Age-Related Changes in mRNA Levels of Hepatic Transporters, Cytochrome P450 and UDP-Glucuronosyltransferase in Female Rats. Eur. J. Drug Metab. Pharmacokinet. 2015, 40, 239–244. [Google Scholar] [CrossRef]
  214. Lerma, E.V. Anatomic and Physiologic Changes of the Aging Kidney. Clin. Geriatr. Med. 2009, 25, 325–329. [Google Scholar] [CrossRef] [PubMed]
  215. Han, B.H.; Sherman, S.; Mauro, P.M.; Martins, S.S.; Rotenberg, J.; Palamar, J.J. Demographic Trends among Older Cannabis Users in the United States, 2006–2013. Addiction 2017, 112, 516–525. [Google Scholar] [CrossRef] [PubMed]
  216. Han, B.H.; Palamar, J.J. Trends in Cannabis Use Among Older Adults in the United States, 2015–2018. JAMA Intern. Med. 2020, 180, 609–611. [Google Scholar] [CrossRef] [PubMed]
  217. Suzuki, S.; Fleig, A.; Penner, R. CBGA Ameliorates Inflammation and Fibrosis in Nephropathy. Sci. Rep. 2023, 13, 6341. [Google Scholar] [CrossRef] [PubMed]
  218. Baban, B.; Hoda, N.; Malik, A.; Khodadadi, H.; Simmerman, E.; Vaibhav, K.; Mozaffari, M.S. Impact of Cannabidiol Treatment on Regulatory T-17 Cells and Neutrophil Polarization in Acute Kidney Injury. Am. J. Physiol. Ren. Physiol. 2018, 315, F1149–F1158. [Google Scholar] [CrossRef] [PubMed]
  219. Worth, H.; O’Hara, D.V.; Agarwal, N.; Collister, D.; Brennan, F.; Smyth, B. Cannabinoids for Symptom Management in Patients with Kidney Failure: A Narrative Review. Clin. J. Am. Soc. Nephrol. 2022, 17, 911–921. [Google Scholar] [CrossRef] [PubMed]
  220. Ho, C.; Martinusen, D.; Lo, C. A Review of Cannabis in Chronic Kidney Disease Symptom Management. Can. J. Kidney Health Dis. 2019, 6, 2054358119828391. [Google Scholar] [CrossRef] [PubMed]
  221. Teixeira-Clerc, F.; Belot, M.-P.; Manin, S.; Deveaux, V.; Cadoudal, T.; Chobert, M.-N.; Louvet, A.; Zimmer, A.; Tordjmann, T.; Mallat, A.; et al. Beneficial Paracrine Effects of Cannabinoid Receptor 2 on Liver Injury and Regeneration. Hepatology 2010, 52, 1046–1059. [Google Scholar] [CrossRef] [PubMed]
  222. Woerdenbag, H.J.; Olinga, P.; Kok, E.A.; Brugman, D.A.P.; van Ark, U.F.; Ramcharan, A.S.; Lebbink, P.W.; Hoogwater, F.J.H.; Knapen, D.G.; de Groot, D.J.A.; et al. Potential, Limitations and Risks of Cannabis-Derived Products in Cancer Treatment. Cancers 2023, 15, 2119. [Google Scholar] [CrossRef]
  223. Blake, A.; Wan, B.A.; Malek, L.; DeAngelis, C.; Diaz, P.; Lao, N.; Chow, E.; O’Hearn, S. A Selective Review of Medical Cannabis in Cancer Pain Management. Ann. Palliat. Med. 2017, 6, S215–S222. [Google Scholar] [CrossRef]
  224. Bar-Sela, G.; Zalman, D.; Semenysty, V.; Ballan, E. The Effects of Dosage-Controlled Cannabis Capsules on Cancer-Related Cachexia and Anorexia Syndrome in Advanced Cancer Patients: Pilot Study. Integr. Cancer Ther. 2019, 18, 153473541988149. [Google Scholar] [CrossRef] [PubMed]
  225. Häuser, W.; Fitzcharles, M.-A.; Radbruch, L.; Petzke, F. Cannabinoids in Pain Management and Palliative Medicine. Dtsch. Ärzteblatt Int. 2017, 114, 627–634. [Google Scholar] [CrossRef] [PubMed]
  226. Hardy, J.; Greer, R.; Huggett, G.; Kearney, A.; Gurgenci, T.; Good, P. Phase IIb Randomized, Placebo-Controlled, Dose-Escalating, Double-Blind Study of Cannabidiol Oil for the Relief of Symptoms in Advanced Cancer (MedCan1-CBD). J. Clin. Oncol. 2023, 41, 1444–1452. [Google Scholar] [CrossRef] [PubMed]
  227. Nathan, R.; Mupamombe, C.T.; Elibol, J.; Case, A.A.; Smith, D.; Hyland, A.; Attwood, K.; Hansen, E.D. Assessing Efficacy and Use Patterns of Medical Cannabis for Symptom Management in Elderly Cancer Patients. Am. J. Hosp. Palliat. Care 2023, 40, 368–373. [Google Scholar] [CrossRef] [PubMed]
  228. Steele, G.; Arneson, T.; Zylla, D. A Comprehensive Review of Cannabis in Patients with Cancer: Availability in the USA, General Efficacy, and Safety. Curr. Oncol. Rep. 2019, 21, 10. [Google Scholar] [CrossRef]
  229. Dixon, T.; Cadenhead, K.S. Cannabidiol versus Placebo as Adjunctive Treatment in Early Psychosis: Study Protocol for Randomized Controlled Trial. Trials 2023, 24, 775. [Google Scholar] [CrossRef]
  230. Leweke, F.M.; Rohleder, C.; Gerth, C.W.; Hellmich, M.; Pukrop, R.; Koethe, D. Cannabidiol and Amisulpride Improve Cognition in Acute Schizophrenia in an Explorative, Double-Blind, Active-Controlled, Randomized Clinical Trial. Front. Pharmacol. 2021, 12, 614811. [Google Scholar] [CrossRef] [PubMed]
  231. Rizkallah, E.; Mongeau-Pérusse, V.; Lamanuzzi, L.; Castenada-Ouellet, S.; Stip, E.; Juteau, L.-C.; Brissette, S.; Bruneau, J.; Dubreucq, S.; Jutras-Aswad, D. Cannabidiol Effects on Cognition in Individuals with Cocaine Use Disorder: Exploratory Results from a Randomized Controlled Trial. Pharmacol. Biochem. Behav. 2022, 216, 173376. [Google Scholar] [CrossRef]
  232. Ortiz, R.; Rueda, S.; Di Ciano, P. Use of Cannabidiol (CBD) for the Treatment of Cognitive Impairment in Psychiatric and Neurological Illness: A Narrative Review. Exp. Clin. Psychopharmacol. 2023, 31, 978–988. [Google Scholar] [CrossRef]
  233. Singh, K.; Bhushan, B.; Chanchal, D.K.; Sharma, S.K.; Rani, K.; Yadav, M.K.; Porwal, P.; Kumar, S.; Sharma, A.; Virmani, T.; et al. Emerging Therapeutic Potential of Cannabidiol (CBD) in Neurological Disorders: A Comprehensive Review. Behav. Neurol. 2023, 2023, 8825358. [Google Scholar] [CrossRef]
  234. Gherzi, M.; Milano, G.; Fucile, C.; Calevo, M.G.; Mancardi, M.M.; Nobili, L.; Astuni, P.; Marini, V.; Barco, S.; Cangemi, G.; et al. Safety and Pharmacokinetics of Medical Cannabis Preparation in a Monocentric Series of Young Patients with Drug Resistant Epilepsy. Complement. Ther. Med. 2020, 51, 102402. [Google Scholar] [CrossRef] [PubMed]
  235. Vermersch, P. Sativex(®) (Tetrahydrocannabinol + Cannabidiol), an Endocannabinoid System Modulator: Basic Features and Main Clinical Data. Expert. Rev. Neurother. 2011, 11, 15–19. [Google Scholar] [CrossRef] [PubMed]
  236. Meng, H.; Johnston, B.; Englesakis, M.; Moulin, D.E.; Bhatia, A. Selective Cannabinoids for Chronic Neuropathic Pain: A Systematic Review and Meta-Analysis. Anesth. Analg. 2017, 125, 1638–1652. [Google Scholar] [CrossRef]
  237. Ren, Y.; Whittard, J.; Higuera-Matas, A.; Morris, C.V.; Hurd, Y.L. Cannabidiol, a Nonpsychotropic Component of Cannabis, Inhibits Cue-Induced Heroin Seeking and Normalizes Discrete Mesolimbic Neuronal Disturbances. J. Neurosci. 2009, 29, 14764–14769. [Google Scholar] [CrossRef]
  238. Hurd, Y.L.; Spriggs, S.; Alishayev, J.; Winkel, G.; Gurgov, K.; Kudrich, C.; Oprescu, A.M.; Salsitz, E. Cannabidiol for the Reduction of Cue-Induced Craving and Anxiety in Drug-Abstinent Individuals with Heroin Use Disorder: A Double-Blind Randomized Placebo-Controlled Trial. Am. J. Psychiatry 2019, 176, 911–922. [Google Scholar] [CrossRef] [PubMed]
  239. Tartakover Matalon, S.; Azar, S.; Meiri, D.; Hadar, R.; Nemirovski, A.; Abu Jabal, N.; Konikoff, F.M.; Drucker, L.; Tam, J.; Naftali, T. Endocannabinoid Levels in Ulcerative Colitis Patients Correlate with Clinical Parameters and Are Affected by Cannabis Consumption. Front. Endocrinol. 2021, 12, 685289. [Google Scholar] [CrossRef]
  240. Couch, D.G.; Cook, H.; Ortori, C.; Barrett, D.; Lund, J.N.; O’Sullivan, S.E. Palmitoylethanolamide and Cannabidiol Prevent Inflammation-Induced Hyperpermeability of the Human Gut In Vitro and In Vivo—A Randomized, Placebo-Controlled, Double-Blind Controlled Trial. Inflamm. Bowel Dis. 2019, 25, 1006–1018. [Google Scholar] [CrossRef] [PubMed]
  241. Lehmann, C.; Fisher, N.B.; Tugwell, B.; Szczesniak, A.; Kelly, M.; Zhou, J. Experimental Cannabidiol Treatment Reduces Early Pancreatic Inflammation in Type 1 Diabetes. Clin. Hemorheol. Microcirc. 2016, 64, 655–662. [Google Scholar] [CrossRef] [PubMed]
  242. Yeshurun, M.; Shpilberg, O.; Herscovici, C.; Shargian, L.; Dreyer, J.; Peck, A.; Israeli, M.; Levy-Assaraf, M.; Gruenewald, T.; Mechoulam, R.; et al. Cannabidiol for the Prevention of Graft-versus-Host-Disease after Allogeneic Hematopoietic Cell Transplantation: Results of a Phase II Study. Biol. Blood Marrow Transpl. 2015, 21, 1770–1775. [Google Scholar] [CrossRef]
  243. Abrams, D.I.; Hilton, J.F.; Leiser, R.J.; Shade, S.B.; Elbeik, T.A.; Aweeka, F.T.; Benowitz, N.L.; Bredt, B.M.; Kosel, B.; Aberg, J.A.; et al. Short-Term Effects of Cannabinoids in Patients with HIV-1 Infection: A Randomized, Placebo-Controlled Clinical Trial. Ann. Intern. Med. 2003, 139, 258–266. [Google Scholar] [CrossRef]
  244. Slawek, D.E.; Arnsten, J.; Sohler, N.; Zhang, C.; Grossberg, R.; Stein, M.; Cunningham, C.O. Daily and Near-Daily Cannabis Use Is Associated with HIV Viral Load Suppression in People Living with HIV Who Use Cocaine. AIDS Care 2021, 33, 1350–1357. [Google Scholar] [CrossRef] [PubMed]
  245. Woolridge, E.; Barton, S.; Samuel, J.; Osorio, J.; Dougherty, A.; Holdcroft, A. Cannabis Use in HIV for Pain and Other Medical Symptoms. J. Pain. Symptom Manag. 2005, 29, 358–367. [Google Scholar] [CrossRef] [PubMed]
  246. Ellis, R.J.; Toperoff, W.; Vaida, F.; van den Brande, G.; Gonzales, J.; Gouaux, B.; Bentley, H.; Atkinson, J.H. Smoked Medicinal Cannabis for Neuropathic Pain in HIV: A Randomized, Crossover Clinical Trial. Neuropsychopharmacology 2009, 34, 672–680. [Google Scholar] [CrossRef] [PubMed]
  247. Abrams, D.I.; Jay, C.A.; Shade, S.B.; Vizoso, H.; Reda, H.; Press, S.; Kelly, M.E.; Rowbotham, M.C.; Petersen, K.L. Cannabis in Painful HIV-Associated Sensory Neuropathy: A Randomized Placebo-Controlled Trial. Neurology 2007, 68, 515–521. [Google Scholar] [CrossRef] [PubMed]
  248. Riggs, P.K.; Vaida, F.; Rossi, S.S.; Sorkin, L.S.; Gouaux, B.; Grant, I.; Ellis, R.J. A Pilot Study of the Effects of Cannabis on Appetite Hormones in HIV-Infected Adult Men. Brain Res. 2012, 1431, 46–52. [Google Scholar] [CrossRef]
  249. Watson, C.W.-M.; Sundermann, E.; Helm, J.; Paolillo, E.W.; Hong, S.; Ellis, R.J.; Letendre, S.; Marcotte, T.D.; Heaton, R.K.; Morgan, E.E.; et al. A Longitudinal Study of Cannabis Use and Risk for Cognitive and Functional Decline among Older Adults with HIV. AIDS Behav. 2023. [Google Scholar] [CrossRef]
Table 1. Summary of cannabidiol (CBD) and delta-9-tetrahydrocannabinol (THC) metabolisms and these cannabinoids as perpetrators of DDIs.
Table 1. Summary of cannabidiol (CBD) and delta-9-tetrahydrocannabinol (THC) metabolisms and these cannabinoids as perpetrators of DDIs.
ComponentCannabinoidsKey Points
EnzymesCBDPhase I (main): CYP3A4, CYP2C9, and CYP2C19 [36];
Phase II (minor): UGT1A9, UGT2B7, and UGT2B17 [38,41].
THCPhase I: CYP3A4, CYP2C9, and CYP2C19 [41].
MetabolitesCBDPrimary: 7-OH-CBD by CYP2C9 and CYP2C19; active [37,78];
Secondary: 7-COOH-CBD, enzyme unidentified; inactive [79].
THCPrimary: 11-OH-THC by CYP2C9 and CYP2C19; active [41];
Secondary: 11-COOH-THC by CYP3A4, CYP2C9, CYP2C19, alcohol dehydrogenases, ALDHs, and aldehyde oxidase; inactive [41,79,83,84].
Perpetrator
of drug interaction
CBDCompetitively inhibits CYP2B6 (Ki,u: 3.4 μM), CYP2C9 (Ki,u: 0.093–1.3 μM), CYP2C19 (Ki,u: 0.050–1.1 μM), CYP2D6 (Ki,u: 0.074 μM), CYP2E1 (Ki,u: 0.021 μM), CYP3A4 (Ki,u: 0.093 μM), and CES1 (Ki,u: 0.091–0.97 μM) in vitro [41];
Time-dependently inhibits CYP1A1 (Ki,u: 5.9 μM), CYP1A2 (Ki,u: 0.11–0.59 μM), and CYP1B1 (Ki,u: 3.1 μM) in vitro [41];
Mixed-type inhibits CYP1A2 (Ki,u: 0.12 μM) in vitro [85];
Also inhibits UGT1A6 (IC50,u: 0.40 μM), UGT1A9 (IC50,u: 0.073–0.12 μM), UGT2B4 (IC50,u: 0.22 μM), and UGT2B7 (IC50,u: 0.82 μM) in vitro [41];
Induces CYP1A2, CYP2B6, and CYP3A4 in vitro, parameters not reported [86].
THCCompetitively inhibits CYP1A2 (Ki,u: 0.090 μM), CYP2B6 (Ki,u: 0.25 μM), CYP2C9 (Ki,u: 0.073 μM), CYP2D6 (Ki,u: 0.11 μM), and CES1 (Ki,u: 0.031–0.54 μM) in vitro [41];
Time-dependently inhibits CYP1A1 (Ki: 1.2 μM) and CYP2A6 (Ki: 0.86 μM) in vitro [41];
Mixed-type inhibits CYP2C19 (Ki,u: 0.056 μM) in vitro [85];
Also inhibits UGT1A9 (IC50,u: 0.33–0.49 μM), UGT2B4 (IC50,u: 0.47 μM), and UGT2B7 (IC50,u: 1.4 μM) in vitro [41].
Additional Factors Body weight; fat distribution/composition; protein binding
CYP, cytochromes P450; UGT, UDP-glucuronosyltransferases; ALDH, aldehyde dehydrogenase; CES1, carboxylesterase 1; DDI, drug–drug interaction; Ki,u, apparent unbound inhibitory constant; IC50,u, 50% inhibitory unbound concentration; Ki, apparent inhibitory constant.
Table 2. Pharmacokinetic parameters for cannabidiol (CBD) in special populations.
Table 2. Pharmacokinetic parameters for cannabidiol (CBD) in special populations.
PopulationDoseAUC0–t (ng·h/mL)Cmax
(ng/mL)
t1/2
(h)
V/F
(L)
Study
Pediatric patients with
epilepsy
10 mg/kg/day, oral173.9
(CV%: 103.3)
59.03
(CV%: 169.4)
31.3
(CV%: 74.9)
-Wheless 2019 [52]
20 mg/kg/day, oral507.1
(CV%: 135.6)
110.5
(CV%: 128.8)
33.5
(CV%: 44.7)
-Wheless 2019 [52]
40 mg/kg/day, oral914.5
(CV%: 126.3)
256.9
(CV%: 136.9)
21.6
(CV%: 48.7)
-Wheless 2019 [52]
12.2 mg/kg/day, oral226.3
(70.5–861.3) 1,2
49.6
(14.4–302.0) 1,2
--Guido 2021 [115]
0.13–5.0 mg/kg, oral-13.1
(2.0–112.7) 1
6.2
(2.8–9.1)
2392
(188–9174) 1
Wang 2020 [116]
Patients with mild
hepatic impairment
200 mg, oral648 (44.2) 3233 (70.5) 315.7 (58.3) 35302 (60.1) 3Taylor 2019 [117]
Patients with moderate
hepatic impairment
200 mg, oral1054 (38.9) 3354 (42.3) 320.5 (39.2) 34668 (40.1) 3Taylor 2019 [117]
Patients with severe
hepatic impairment
200 mg, oral1855 (52.0) 3381 (52.2) 322.1 (44.9) 32437 (70.5) 3Taylor 2019 [117]
Patients with mild
renal impairment
200 mg, oral671 (40.9) 3200 (42.7) 315.5
(CV%: 64.5)
6661
(CV%: 55.5)
Tayo 2020 [118]
Patients with moderate
renal impairment
200 mg, oral530 (74.4) 3172 (85.3) 314.6
(CV%: 46.6)
7778
(CV%: 58.0)
Tayo 2020 [118]
Patients with severe
renal impairment
200 mg, oral532 (32.7) 3155 (40.6) 313.1
(CV%: 41.5)
6016
(CV%: 39.9)
Tayo 2020 [118]
Patients with advanced
cancer
2.5 mg,
oromucosal spray
0.65
(0.49–4.06) 1
0.58
(0.48–2.45) 1
0.72
(0.57–0.86) 1
-Clarke 2022 [119]
7.5 mg,
oromucosal spray
5.96
(1.51–12.15) 1
1.55
(0.62–2.25) 1
1.53
(1.16–7.06) 1
-Clarke 2022 [119]
Adult patients with
epilepsy
200–300 mg, oral0.53
(0.26, (ng·h/mL)/mg) 4
0.03
(0.01, (ng/mL)/mg) 4
38.9 (19) 41515
(1024, L/kg) 4
Birnbaum 2019 [120]
Patients with multiple
sclerosis
5 mg, oral9.44
(CI: 8.5–10.4) 5
1.33
(CI: 1.20–1.46)
-3912
(CI: 3565–4260)
Hansen 2024 [121]
10 mg, oral19.02
(CI: 17.0–21.0) 5
2.67
(CI: 2.39–2.95)
-3781
(CI: 3341–4222)
Hansen 2024 [121]
15 mg, oral32.35
(CI: 31.6–33.1) 5
4.54
(CI: 4.44–4.65)
-3435
(CI: 3345–3525)
Hansen 2024 [121]
Patients with Crohn’s
disease
7.5 mg, oral40.32
(SD: 25.65) 5
8.1
(SD: 5.4)
0.65
(SD: 0.42)
-Naftali 2021 [122]
If not labeled, data are presented as mean (range). AUC0–t, area under the plasma concentration–time curve from time zero to last measurable concentration; AUC0–inf, area under the curve to infinity; Cmax, maximum measured plasma concentration; t1/2, terminal (elimination) half-life; V/F, apparent volume of distribution; CV, coefficient of variation; CI, 95% confidence interval; SD, standard deviation; -, data not reported. 1 Median (range). 2 At steady state. 3 Geometric mean (Geometric CV%). 4 Geometric mean (SD). 5 AUC0–inf.
Table 3. Pharmacokinetic parameters for delta-9-tetrahydrocannabinol (THC) in special populations.
Table 3. Pharmacokinetic parameters for delta-9-tetrahydrocannabinol (THC) in special populations.
PopulationDoseAUC0–t (ng·h/mL)Cmax
(ng/mL)
t1/2
(h)
V/F
(L)
Study
Healthy older adults3 mg-1.42
(0.53–3.48)
--Ahmed 2014 [123]
5 mg-3.15
(1.54–6.95)
--Ahmed 2014 [123]
6.5 mg-4.57
(2.11–8.65)
--Ahmed 2014 [123]
Older adults with dementia0.75 mg0.88
(CV%: 124)
0.41
(CV%: 138)
5.08
(CV%: 39)
-Ahmed 2015 [124]
1.5 mg2.01
(CV%: 136)
1.01
(CV%: 112)
5.06
(CV%: 37)
-Ahmed 2015 [124]
Patients with advanced cancer2.5 mg
oromucosal spray
1.71
(1.11–6.61) 1
1.31
(0.76–2.94) 1
0.94
(0.75–1.14) 1
-Clarke 2022 [119]
7.5 mg
oromucosal spray
8.26
(2.67–11.72) 1
2.35
(1.09–3.19) 1
1.39
(1.30–2.88) 1
-Clarke 2022 [119]
Patients with multiple
sclerosis
2.5 mg oral3.23
(CI: 2.7–3.7)
0.57
(CI: 0.50–0.63)
-3173
(CI: 2753–3594)
Hansen 2024 [121]
5 mg oral7.03
(CI: 6.6–7.5)
1.21
(CI: 1.15–1.26)
-2906
(CI: 2728–3084)
Hansen 2024 [121]
7.5 mg oral10.81
(CI: 10.5–11.1)
1.85
(CI: 1.81–1.88)
-2831
(CI: 2754–2909)
Hansen 2024 [121]
Patients with neuropathic pain0.5 mg inhaled5
(SD: 2.4) 2
14.3
(SD: 7.7)
--Almog 2020 [125]
1.0 mg inhaled12.8
(SD: 5.5) 2
33.8
(SD: 25.7)
--Almog 2020 [125]
3.08 mg inhaled10.1
(SD: 3.3)
38
(SD: 10)
--Eisenberg 2014 [126]
Patients with Crohn’s disease2 mg oral10.7
(SD: 2.2)
3.0
(SD: 2.1)
0.55
(SD: 0.27)
-Naftali 2021 [122]
If not labeled, data are presented as mean (range). AUC0–t, area under the plasma concentration–time curve from time zero to last measurable concentration; AUC0–inf, area under the curve to infinity; Cmax, maximum measured plasma concentration; t1/2, terminal (elimination) half-life; V/F, apparent volume of distribution; CV, coefficient of variation; CI, 95% confidence interval; SD, standard deviation; -, data not reported. 1 Median (range). 2 AUC0–inf.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Qian, L.; Beers, J.L.; Jackson, K.D.; Zhou, Z. CBD and THC in Special Populations: Pharmacokinetics and Drug–Drug Interactions. Pharmaceutics 2024, 16, 484. https://doi.org/10.3390/pharmaceutics16040484

AMA Style

Qian L, Beers JL, Jackson KD, Zhou Z. CBD and THC in Special Populations: Pharmacokinetics and Drug–Drug Interactions. Pharmaceutics. 2024; 16(4):484. https://doi.org/10.3390/pharmaceutics16040484

Chicago/Turabian Style

Qian, Lixuan, Jessica L. Beers, Klarissa D. Jackson, and Zhu Zhou. 2024. "CBD and THC in Special Populations: Pharmacokinetics and Drug–Drug Interactions" Pharmaceutics 16, no. 4: 484. https://doi.org/10.3390/pharmaceutics16040484

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop