Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (49)

Search Parameters:
Keywords = cannabidiol pharmacokinetics

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
39 pages, 2934 KiB  
Review
Phytocannabinoids as Novel SGLT2 Modulators for Renal Glucose Reabsorption in Type 2 Diabetes Management
by Raymond Rubianto Tjandrawinata, Dante Saksono Harbuwono, Sidartawan Soegondo, Nurpudji Astuti Taslim and Fahrul Nurkolis
Pharmaceuticals 2025, 18(8), 1101; https://doi.org/10.3390/ph18081101 - 24 Jul 2025
Viewed by 485
Abstract
Background: Sodium–glucose cotransporter 2 (SGLT2) inhibitors have transformed type 2 diabetes mellitus (T2DM) management by promoting glucosuria, lowering glycated hemoglobin (HbA1c), blood pressure, and weight; however, their use is limited by genitourinary infections and ketoacidosis. Phytocannabinoids—bioactive compounds from Cannabis sativa—exhibit multi-target [...] Read more.
Background: Sodium–glucose cotransporter 2 (SGLT2) inhibitors have transformed type 2 diabetes mellitus (T2DM) management by promoting glucosuria, lowering glycated hemoglobin (HbA1c), blood pressure, and weight; however, their use is limited by genitourinary infections and ketoacidosis. Phytocannabinoids—bioactive compounds from Cannabis sativa—exhibit multi-target pharmacology, including interactions with cannabinoid receptors, Peroxisome Proliferator-Activated Receptors (PPARs), Transient Receptor Potential (TRP) channels, and potentially SGLT2. Objective: To evaluate the potential of phytocannabinoids as novel modulators of renal glucose reabsorption via SGLT2 and to compare their efficacy, safety, and pharmacological profiles with synthetic SGLT2 inhibitors. Methods: We performed a narrative review encompassing the following: (1) the molecular and physiological roles of SGLT2; (2) chemical classification, natural sources, and pharmacokinetics/pharmacodynamics of major phytocannabinoids (Δ9-Tetrahydrocannabinol or Δ9-THC, Cannabidiol or CBD, Cannabigerol or CBG, Cannabichromene or CBC, Tetrahydrocannabivarin or THCV, and β-caryophyllene); (3) in silico docking and drug-likeness assessments; (4) in vitro assays of receptor binding, TRP channel modulation, and glucose transport; (5) in vivo rodent models evaluating glycemic control, weight change, and organ protection; (6) pilot clinical studies of THCV and case reports of CBD/BCP; (7) comparative analysis with established synthetic inhibitors. Results: In silico studies identify high-affinity binding of several phytocannabinoids within the SGLT2 substrate pocket. In vitro, CBG and THCV modulate SGLT2-related pathways indirectly via TRP channels and CB receptors; direct IC50 values for SGLT2 remain to be determined. In vivo, THCV and CBD demonstrate glucose-lowering, insulin-sensitizing, weight-reducing, anti-inflammatory, and organ-protective effects. Pilot clinical data (n = 62) show that THCV decreases fasting glucose, enhances β-cell function, and lacks psychoactive side effects. Compared to synthetic inhibitors, phytocannabinoids offer pleiotropic benefits but face challenges of low oral bioavailability, polypharmacology, inter-individual variability, and limited large-scale trials. Discussion: While preclinical and early clinical data highlight phytocannabinoids’ potential in SGLT2 modulation and broader metabolic improvement, their translation is impeded by significant challenges. These include low oral bioavailability, inconsistent pharmacokinetic profiles, and the absence of standardized formulations, necessitating advanced delivery system development. Furthermore, the inherent polypharmacology of these compounds, while beneficial, demands comprehensive safety assessments for potential off-target effects and drug interactions. The scarcity of large-scale, well-controlled clinical trials and the need for clear regulatory frameworks remain critical hurdles. Addressing these aspects is paramount to fully realize the therapeutic utility of phytocannabinoids as a comprehensive approach to T2DM management. Conclusion: Phytocannabinoids represent promising multi-target agents for T2DM through potential SGLT2 modulation and complementary metabolic effects. Future work should focus on pharmacokinetic optimization, precise quantification of SGLT2 inhibition, and robust clinical trials to establish efficacy and safety profiles relative to synthetic inhibitors. Full article
Show Figures

Graphical abstract

19 pages, 1882 KiB  
Article
Pharmacokinetics of Cannabidiol in Rat Brain Tissue After Single-Dose Administration of Different Formulations
by Zuzana Binova, Frantisek Benes, Marie Zlechovcova, Matej Maly, Petr Kastanek, Monika Cahova, Milena Stranska and Jana Hajslova
Molecules 2025, 30(13), 2676; https://doi.org/10.3390/molecules30132676 - 20 Jun 2025
Viewed by 441
Abstract
Cannabidiol (CBD), a phytocannabinoid commonly isolated from chemotype III Cannabis sativa plants, is known for its therapeutic potential. However, comprehensive information on its bioavailability is still lacking. The key objective of this study was to investigate the impact of specific formulations on CBD [...] Read more.
Cannabidiol (CBD), a phytocannabinoid commonly isolated from chemotype III Cannabis sativa plants, is known for its therapeutic potential. However, comprehensive information on its bioavailability is still lacking. The key objective of this study was to investigate the impact of specific formulations on CBD delivery to the site of action and, in particular, the brain of experimental animals. As brain tissue is an extremely complex matrix, a highly sensitive method employing liquid chromatography–tandem mass spectrometry (LC-MS/MS) had to be implemented. To make it applicable for multiple analytes, the method was validated for 17 other phytocannabinoids and selected metabolites. Using this method, a pharmacokinetic study was conducted on 200 brain samples collected from rats that had been administered various CBD formulations (carriers) via oral gavage. The peak concentration in brain occurred within 1–2 h; notably, the highest was reached with carriers containing triacylglycerols with the shortest fatty acid chains (caprylic/capric). In addition to the parent compound, 7-hydroxy-cannabidiol and 7-carboxy-cannabidiol were detected, confirming rapid post-administration metabolism. Overall, this research enhances understanding of CBD distribution in the brain and underscores the impact of specific formulations on its bioavailability, offering insights into optimizing CBD-based therapies to be both effective and ‘patient-friendly’. Full article
(This article belongs to the Special Issue Recent Advances in Cannabis and Hemp Research)
Show Figures

Graphical abstract

14 pages, 515 KiB  
Article
Pharmacokinetic Characteristics of a Single Cannabidiol Dose in Oil and Treat Forms and Health Impacts After 30 Days of Administration in Dogs
by Phattharakan Kamutchat, Sasithorn Limsuwan, Nattaya Leewichit and Natthasit Tansakul
Animals 2025, 15(10), 1470; https://doi.org/10.3390/ani15101470 - 19 May 2025
Viewed by 874
Abstract
Cannabidiol (CBD) has garnered significant interest in veterinary therapeutics, yet the pharmacokinetic and safety profiles of its various formulations remain incompletely characterized. This study compared the pharmacokinetics (PK) and health effects of CBD administered as oil (OG, 5 mg/kg) and treats (TG, 50 [...] Read more.
Cannabidiol (CBD) has garnered significant interest in veterinary therapeutics, yet the pharmacokinetic and safety profiles of its various formulations remain incompletely characterized. This study compared the pharmacokinetics (PK) and health effects of CBD administered as oil (OG, 5 mg/kg) and treats (TG, 50 mg) in 16 healthy mixed-breed dogs over 30 days. Plasma CBD concentrations were measured using liquid chromatography–tandem mass spectrometry (LC-MS/MS), and the PK parameters were analyzed using non-compartmental methods. The CBD-infused rice bran oil formulation (OG) achieved a significantly higher dose-normalized maximum plasma concentration (Cmax, 58.40 vs. 21.29 kg·ng/mL/mg) and area under the curve (AUC0-inf, 305.85 vs. 141.75 h·kg·ng/mL/mg) compared to the treats (TG). The treat formulation exhibited relative reductions in bioavailability, with AUC and Cmax values approximately 2.2- and 2.7-fold lower than the oil group. The terminal half-life (~9.66 h OG vs. 8.52 h TG) and time to peak (2.38 h OG vs. 3.63 h TG) did not differ significantly. CBD accumulation occurred with repeated dosing but declined rapidly post-cessation. The hematological and biochemical analyses revealed no clinically adverse effects, though minor erythrocyte and eosinophil fluctuations were noted. The oil formulation demonstrated superior absorption, while both forms were well-tolerated. These findings highlight the impact of formulation on CBD absorption and support further research into optimized delivery methods for veterinary applications. Full article
(This article belongs to the Section Companion Animals)
Show Figures

Figure 1

15 pages, 1838 KiB  
Article
A Preliminary Evaluation of the Comparative Efficacy of Gel-Based and Oil-Based CBD on Hematologic and Biochemical Responses in Dogs
by Wassana Puttharaksa, Rangsun Charoensook, Rongdej Tungtrakanpoung, Niramon Hoidokhom, Saowaluk Rungchang, Bertram Brenig and Sonthaya Numthuam
Vet. Sci. 2025, 12(4), 342; https://doi.org/10.3390/vetsci12040342 - 7 Apr 2025
Cited by 1 | Viewed by 955
Abstract
Cannabidiol (CBD) has gained popularity in veterinary medicine for its potential to alleviate stress, pain, and inflammation in dogs. However, its oral administration is limited by hydrophobicity, variable absorption, and extensive first-pass metabolism, which requires optimized delivery methods to enhance efficacy. This study [...] Read more.
Cannabidiol (CBD) has gained popularity in veterinary medicine for its potential to alleviate stress, pain, and inflammation in dogs. However, its oral administration is limited by hydrophobicity, variable absorption, and extensive first-pass metabolism, which requires optimized delivery methods to enhance efficacy. This study investigated the effects of daily oral supplementation of CBD oil and CBD gel (each at 4 mg/kg), compared to a placebo, over 14 days in shelter dogs subjected to solitary confinement-induced stress. Both CBD formulations appeared safe under the study conditions, with no adverse effects on hematological and biochemical parameters. Post-stress cortisol levels were significantly lower in CBD-treated groups compared to controls, with CBD-infused gel showing a pattern toward greater attenuation. Multivariate analysis revealed distinct blood profile shifts in CBD-treated dogs, with PCA loadings indicating associations between CBD supplementation and lymphocyte percentages and IgG levels. These findings support gel-based CBD as a promising strategy for stress modulation in dogs. Further studies should explore its pharmacokinetics and long-term immune effects to optimize veterinary applications. Full article
(This article belongs to the Section Veterinary Biomedical Sciences)
Show Figures

Figure 1

28 pages, 6588 KiB  
Article
Formulation and Evaluation of Solid Self-Nanoemulsifying Drug Delivery System of Cannabidiol for Enhanced Solubility and Bioavailability
by Fengying Wu, Qing Ma, Guanghui Tian, Kaixian Chen, Rulei Yang and Jingshan Shen
Pharmaceutics 2025, 17(3), 340; https://doi.org/10.3390/pharmaceutics17030340 - 6 Mar 2025
Cited by 2 | Viewed by 2521
Abstract
Background/Objectives: This study aims to develop a solid self-nanoemulsifying drug delivery system (SNEDDS) to enhance the solubility and oral bioavailability of cannabidiol (CBD). Methods: According to the solubility of CBD and pseudo-ternary phase diagrams of the different ingredients, an oil (medium-chain triglyceride, MCT), [...] Read more.
Background/Objectives: This study aims to develop a solid self-nanoemulsifying drug delivery system (SNEDDS) to enhance the solubility and oral bioavailability of cannabidiol (CBD). Methods: According to the solubility of CBD and pseudo-ternary phase diagrams of the different ingredients, an oil (medium-chain triglyceride, MCT), mixed surfactants (Labrasol, Tween 80), and a co-surfactant (Transcutol) were selected for the SNEDDS. CBD-loaded SNEDDS formulations were prepared and characterized. The optimal SNEDDS was converted into solid SNEDDS powders via solid carrier adsorption and spray drying techniques. Various evaluations including flowability, drug release, self-emulsifying capacity, X-ray diffraction (XRD), differential scanning calorimetry (DSC), Fourier transform infrared spectroscopy (FTIR), morphology, and pharmacokinetic characteristics were conducted. Subsequently, the solid powders with fillers, disintegrants, and lubricants were added to the capsules for accelerated stability testing. Results: The investigations showed that the two S-SNEDDS formulations improved the CBD’s solubility and in vitro drug release, with good storage stability. The pharmacokinetic data of Sprague Dawley rats indicated that a single oral dose of L-SNEDDS and spray drying SNEDDS led to a quicker absorption and a higher Cmax of CBD compared to the two oil-based controls (CBD-sesame oil (similar to Epidiolex®) and CBD-MCT), which is favorable for the application of CBD products. Conclusions: SNEDDS is a prospective strategy for enhancing the solubility and oral bioavailability of CBD, and solid SNEDDS offers flexibility for developing more CBD-loaded solid formulations. Moreover, SNEDDS provides new concepts and methods for other poorly water-soluble drugs. Full article
(This article belongs to the Section Physical Pharmacy and Formulation)
Show Figures

Graphical abstract

32 pages, 1799 KiB  
Review
An Overview of the Potential for Pharmacokinetic Interactions Between Drugs and Cannabis Products in Humans
by Dolly Andrea Caicedo, Clara Pérez-Mañá, Magí Farré and Esther Papaseit
Pharmaceutics 2025, 17(3), 319; https://doi.org/10.3390/pharmaceutics17030319 - 1 Mar 2025
Cited by 2 | Viewed by 7294
Abstract
Cannabis is the most commonly used illicit substance worldwide. Recent years have seen an increase in cannabis consumption, and with new approvals and therapeutic indications, there are challenges in minimizing the risks and interactions between cannabis-based products, cannabis prescription drugs, other approved prescription [...] Read more.
Cannabis is the most commonly used illicit substance worldwide. Recent years have seen an increase in cannabis consumption, and with new approvals and therapeutic indications, there are challenges in minimizing the risks and interactions between cannabis-based products, cannabis prescription drugs, other approved prescription drugs, and other substances of abuse. Thus, identifying the enzymes metabolizing cannabinoid drugs and their relationship with other prescription drugs is crucial for understanding the potential interactions and effects of their simultaneous use. This article offers a comprehensive review of cannabis and the pharmacokinetic interactions between cannabis products, cannabis prescription drugs, and other approved prescription drugs, as well as other substances of abuse. It also compiles existing evidence of these interactions and describes the clinical outcomes associated with the inhibition or induction of various enzymes. Full article
(This article belongs to the Topic Cannabis, Cannabinoids and Its Derivatives)
Show Figures

Figure 1

21 pages, 754 KiB  
Review
Pharmacokinetics of Non-Psychotropic Phytocannabinoids
by Mariana Lacerda, Andreia Carona, Sara Castanheira, Amílcar Falcão, Joana Bicker and Ana Fortuna
Pharmaceutics 2025, 17(2), 236; https://doi.org/10.3390/pharmaceutics17020236 - 12 Feb 2025
Cited by 3 | Viewed by 1701
Abstract
Cannabinoids are widely recognized for their potential therapeutic effects, making them significant and valuable candidates for medical research and applications across various fields. This review aims to analyze the pharmacokinetics of Cannabidiol (CBD), Cannabigerol (CBG), and Cannabichromene (CBC), along with their corresponding acidic [...] Read more.
Cannabinoids are widely recognized for their potential therapeutic effects, making them significant and valuable candidates for medical research and applications across various fields. This review aims to analyze the pharmacokinetics of Cannabidiol (CBD), Cannabigerol (CBG), and Cannabichromene (CBC), along with their corresponding acidic forms, Cannabidiolic acid (CBDA), Cannabigerolic acid (CBGA), and Cannabichromenic acid (CBCA). Among these cannabinoids, CBD is the most extensively studied. Nevertheless, research involving all the mentioned cannabinoids has shown that their pharmacokinetic parameters are highly variable, depending significantly on factors such as dose, formulation, route of administration, and diet. Furthermore, challenges such as brain penetration and first-pass metabolism have been highlighted. In conclusion, this review demonstrates significant progress in understanding the pharmacokinetics of non-psychotropic cannabinoids. However, it also underscores the need for further research, particularly on CBG, CBC, and their respective acidic forms, with the most significant gap being in clinical investigations. Expanding these studies is essential to facilitate their optimized use in medical treatments. Full article
Show Figures

Figure 1

40 pages, 4017 KiB  
Review
Utilization of Cannabidiol in Post-Organ-Transplant Care
by Sachiko Koyama, Jumar Etkins, Joshua Jun, Matthew Miller, Gerald C. So, Debora L. Gisch and Michael T. Eadon
Int. J. Mol. Sci. 2025, 26(2), 699; https://doi.org/10.3390/ijms26020699 - 15 Jan 2025
Viewed by 2013
Abstract
Cannabidiol (CBD) is one of the major phytochemical constituents of cannabis, Cannabis sativa, widely recognized for its therapeutic potential. While cannabis has been utilized for medicinal purposes since ancient times, its psychoactive and addictive properties led to its prohibition in 1937, with [...] Read more.
Cannabidiol (CBD) is one of the major phytochemical constituents of cannabis, Cannabis sativa, widely recognized for its therapeutic potential. While cannabis has been utilized for medicinal purposes since ancient times, its psychoactive and addictive properties led to its prohibition in 1937, with only the medical use being reauthorized in 1998. Unlike tetrahydrocannabinol (THC), CBD lacks psychoactive and addictive properties, yet the name that suggests its association with cannabis has significantly contributed to its public visibility. CBD exhibits diverse pharmacological properties, most notably anti-inflammatory effects. Additionally, it interacts with key drug-metabolizing enzyme families, including cytochrome P450 (CYP) and uridine 5′-diphospho-glucuronosyltransferase (UGT), which mediate phase I and phase II metabolism, respectively. By binding to these enzymes, CBD can inhibit the metabolism of co-administered drugs, which can potentially enhance their toxicity or therapeutic effects. Mild to moderate adverse events associated with CBD use have been reported. Advances in chemical formulation techniques have recently enabled strategies to minimize these effects. This review provides an overview of CBD, covering its historical background, recent clinical trials, adverse event profiles, and interactions with molecular targets such as receptors, channels, and enzymes. We particularly emphasize the mechanisms underlying its anti-inflammatory effects and interaction with drugs relevant to organ transplantation. Finally, we explore recent progress in the chemical formulation of CBD in order to enhance its bioavailability, which will enable decreasing the dose to use and increase its safety and efficacy. Full article
(This article belongs to the Special Issue Recent Research of Phytochemicals in Health and Disease)
Show Figures

Figure 1

14 pages, 2279 KiB  
Article
Evaluation of the Drug–Drug Interaction Potential of Cannabidiol Against UGT2B7-Mediated Morphine Metabolism Using Physiologically Based Pharmacokinetic Modeling
by Shelby Coates, Keti Bardhi, Bhagwat Prasad and Philip Lazarus
Pharmaceutics 2024, 16(12), 1599; https://doi.org/10.3390/pharmaceutics16121599 - 16 Dec 2024
Cited by 2 | Viewed by 1705
Abstract
Background: Morphine is a commonly prescribed opioid analgesic used to treat chronic pain. Morphine undergoes glucuronidation by UDP-glucuronosyltransferase (UGT) 2B7 to form morphine-3-glucuronide and morphine-6-glucuronide. Morphine is the gold standard for chronic pain management and has a narrow therapeutic index. Reports have shown [...] Read more.
Background: Morphine is a commonly prescribed opioid analgesic used to treat chronic pain. Morphine undergoes glucuronidation by UDP-glucuronosyltransferase (UGT) 2B7 to form morphine-3-glucuronide and morphine-6-glucuronide. Morphine is the gold standard for chronic pain management and has a narrow therapeutic index. Reports have shown that chronic pain patients have increasingly used other supplements to treat their chronic pain, including cannabidiol (CBD). Up to 50% of chronic pain patients report that they co-use cannabis with their prescribed opioid for pain management, including morphine. Previous work has shown that cannabidiol is a potent inhibitor of UGT2B7, including morphine-mediated metabolism. Co-use of morphine and CBD may result in unwanted drug–drug interactions (DDIs). Methods: Using available physiochemical and clinical parameters, morphine and CBD physiologically based pharmacokinetic (PBPK) models were developed and validated in both healthy and cirrhotic populations. Models for the two populations were then combined to predict the severity and clinical relevance of the potential DDIs during coadministration of both morphine and CBD in both healthy and hepatic-impaired virtual populations. Results: The predictive DDI model suggests that a ~5% increase in morphine exposure is to be expected in healthy populations. A similar increase in exposure of morphine is predicted in severe hepatic-impaired populations with an increase of ~10. Conclusions: While these predicted increases in morphine exposure are below the Food and Drug Administration’s cutoff (1.25-fold increase), morphine has a narrow therapeutic index and a 5–10% increase in exposure may be clinically relevant. Future clinical studies are needed to fully characterize the clinical relevance of morphine-related DDIs. Full article
Show Figures

Figure 1

19 pages, 5538 KiB  
Article
Novel Lipid Formulation Increases Absorption of Oral Cannabidiol (CBD)
by Edward Chesney, Ndabezinhle Mazibuko, Dominic Oliver, Amedeo Minichino, Ayşe Doğa Lamper, Lucy Chester, Thomas J. Reilly, Millie Lloyd, Matilda Kråkström, Alex M. Dickens, Matej Orešič, Eric Lynch, Gregory Stoloff, Mitul A. Mehta and Philip McGuire
Pharmaceutics 2024, 16(12), 1537; https://doi.org/10.3390/pharmaceutics16121537 - 1 Dec 2024
Cited by 1 | Viewed by 2173
Abstract
Background: Cannabidiol (CBD) is an approved treatment for childhood epilepsies and a candidate treatment for several other CNS disorders. However, it has poor oral bioavailability. We investigated the effect of a novel lipid formulation on its absorption in humans and on its [...] Read more.
Background: Cannabidiol (CBD) is an approved treatment for childhood epilepsies and a candidate treatment for several other CNS disorders. However, it has poor oral bioavailability. We investigated the effect of a novel lipid formulation on its absorption in humans and on its tissue distribution in mice. Methods: In a double-blind crossover study in fasting healthy volunteers, we compared the pharmacokinetics of a single dose of 1000 mg of CBD in the lipid formulation and in a powder formulation (ClinicalTrials.gov: NCT05032807). In a second study, male CD1 mice were administered CBD in either the lipid formulation or dissolved in water, via oral gavage (n = 1 per timepoint). The tissue distribution of CBD was assessed using matrix-assisted laser desorption/ionization mass spectrometric imaging. Results: Plasma exposure (AUC0–48) of CBD was nine times greater for the lipid formulation than the powder formulation (611.1 ng·h/mL [coefficient of variation {CV%}: 104.6] and 66.8 ng·h/mL [CV%: 50.7], respectively). With the powder formulation, the AUC0–48 was related to the concentration of specific gastrointestinal bacteria and bile acids. These associations were attenuated with the lipid formulation. In the animal study, after treatment with the lipid formulation, measurable concentrations of CBD were identified in all organs. For the aqueous formulation, tissue concentrations of CBD were below the limit of quantification. Conclusions: Administering oral CBD in a lipid formulation was associated with an increase in its gastrointestinal absorption, as well as an attenuation of the relationship between its absorption and features of the gut microbiome. Full article
Show Figures

Figure 1

15 pages, 2722 KiB  
Article
LC-MS/MS-Based Concurrent Quantification of Cannabidiol and Melatonin in Mouse Plasma to Elucidate Complex PK Interactions
by Mengran Wang, Wenpeng Zhang, Xia Wu, Lingchao Wang, Cong Li, Chunyan Liu and Xiaomei Zhuang
Pharmaceutics 2024, 16(12), 1511; https://doi.org/10.3390/pharmaceutics16121511 - 25 Nov 2024
Viewed by 1349
Abstract
Objective: This study aimed to develop a quantitative analytical method for the simultaneous determination of cannabidiol (CBD) and melatonin (MT) in mouse plasma using the protein precipitation method coupled with LC-MS/MS. Additionally, this study sought to investigate the impact of CBD on the [...] Read more.
Objective: This study aimed to develop a quantitative analytical method for the simultaneous determination of cannabidiol (CBD) and melatonin (MT) in mouse plasma using the protein precipitation method coupled with LC-MS/MS. Additionally, this study sought to investigate the impact of CBD on the pharmacokinetics of MT in mice using this method. Methods: Mouse plasma samples were precipitated with acetonitrile and analyzed using a Kromasil 100-5-C8 (2.1 × 50 mm) column. Following a single administration, thirty male ICR mice were randomly assigned to five groups: MT 2 mg/kg intravenously (i.v.), MT 10 mg/kg orally (p.o.), MT + CBD (10 + 10) mg/kg p.o., MT + CBD (10 + 40) mg/kg p.o., and MT 10 mg/kg p.o. followed by CBD 2 mg/kg i.v. Pharmacokinetic parameters were calculated using a non-compartmental model and analyzed to investigate the interactions of CBD with MT. Results: The calibration curves for CBD and MT were linear over the range of 2 to 1000 ng/mL. Co-administration of a high dose of CBD (40 mg/kg) orally reduced the Cmax of MT (10 mg/kg) to 57% of the control, while the area under the curve from 0.5 to 8 h (AUC(0.5–8h)) was 2.85-fold that of the MT-only group. When CBD (2 mg/kg) was administered intravenously alongside MT orally, the AUC(0.5–8h) was 1.54 times that of MT given orally alone. The AUC of CBD was positively correlated with the AUC of the distribution and elimination phases of MT, while the Cmax of CBD negatively correlated with the Cmax of MT. Conclusions: The developed LC-MS/MS method is robust and suitable for pharmacokinetic studies involving CBD and MT. The in vivo effects of CBD on MT pharmacokinetics are complex. High oral doses of CBD inhibit both the intestinal absorption and metabolic clearance of MT, resulting in a more smooth PK profile. Full article
Show Figures

Figure 1

14 pages, 1714 KiB  
Article
Structure–Tissue Exposure/Selectivity Relationship (STR) on Carbamates of Cannabidiol
by Sheng Wang, Jian-Guo Yang, Kuanrong Rong, Huan-Huan Li, Chengyao Wu and Wenjian Tang
Int. J. Mol. Sci. 2024, 25(22), 11888; https://doi.org/10.3390/ijms252211888 - 5 Nov 2024
Viewed by 996
Abstract
The structure–tissue exposure/selectivity relationship (STR) aids in lead optimization to improve drug candidate selection and balance clinical dose, efficacy, and toxicity. In this work, butyrocholinesterase (BuChE)-targeted cannabidiol (CBD) carbamates were used to study the STR in correlation with observed efficacy/toxicity. CBD carbamates with [...] Read more.
The structure–tissue exposure/selectivity relationship (STR) aids in lead optimization to improve drug candidate selection and balance clinical dose, efficacy, and toxicity. In this work, butyrocholinesterase (BuChE)-targeted cannabidiol (CBD) carbamates were used to study the STR in correlation with observed efficacy/toxicity. CBD carbamates with similar structures and same molecular target showed similar/different pharmacokinetics. L2 and L4 had almost same plasma exposure, which was not correlated with their exposure in the brain, while tissue exposure/selectivity was correlated with efficacy/safety. Structural modifications of CBD carbamates not only changed drug plasma exposure, but also altered drug tissue exposure/selectivity. The secondary amine of carbamate can be metabolized into CBD, while the tertiary amine is more stable. Absorption, distribution, metabolism, excretion, and toxicity (ADMET) parameters can be used to predict STR. Therefore, STR can alter drug tissue exposure/selectivity in normal tissues, impacting efficacy/toxicity. The drug optimization process should balance the structure–activity relationship (SAR) and STR of drug candidates for improving clinical trials. Full article
(This article belongs to the Section Molecular Informatics)
Show Figures

Graphical abstract

23 pages, 312 KiB  
Review
Drug–Cannabinoid Interactions in Selected Therapeutics for Symptoms Associated with Epilepsy, Autism Spectrum Disorder, Cancer, Multiple Sclerosis, and Pain
by Maria G. Campos, Maria China, Mariana Cláudio, Miguel Capinha, Rita Torres, Simão Oliveira and Ana Fortuna
Pharmaceuticals 2024, 17(5), 613; https://doi.org/10.3390/ph17050613 - 10 May 2024
Cited by 3 | Viewed by 4377
Abstract
Clinical practice entails a translation of research that assists in the use of scientific data and therapeutic evidence for the benefit of the patient. This review critically summarizes the potential impact of cannabinoids in conjunction with other drugs when associated with treatments for [...] Read more.
Clinical practice entails a translation of research that assists in the use of scientific data and therapeutic evidence for the benefit of the patient. This review critically summarizes the potential impact of cannabinoids in conjunction with other drugs when associated with treatments for epilepsy, autism spectrum disorder, cancer, multiple sclerosis, and chronic pain. In these associations, potential drug interactions may occur and alter the predicted clinical results. Therefore, the potential for drug interactions must always be assessed to avoid therapeutic failures and/or increased side effects. Some effects may be additive, synergistic, or antagonistic, but changes in absorption, distribution, metabolism, particularly through cytochrome P450 (CYP) isoenzymes (e.g., CYP2C9 and CYP3A4), and excretion may also occur. For example, the combination of cannabis-derived compounds and the antifungal drug ketoconazole, a CYP3A4 inhibitor, increases the plasma concentration of Δ-9-tetrahydrocannabinol (THC) and cannabidiol (CBD). In contrast, rifampicin, a CYP3A4 inducer, stands out for reducing plasma THC levels by approximately 20–40% and 50% to 60% for CBD. Other CYP3A4 inhibitors and inducers are likely to have a similar effect on plasma concentrations if co-administered. Pharmacokinetic interactions with anticonvulsant medications have also been reported, as have pharmacodynamic interactions between cannabinoids and medications with sympathomimetic effects (e.g., tachycardia, hypertension), central nervous system depressants (e.g., drowsiness, ataxia), and anticholinergics (e.g., tachycardia and somnolence). Although further studies are still pending, there is currently clinical evidence supporting drug interactions with cannabinoids, requiring doctors to evaluate the risk of drug combinations with cannabinoids and vice versa. The tables provided here were designed to facilitate the identification of biorelevant interactions that may compromise therapeutic efficacy and toxicity. Full article
27 pages, 483 KiB  
Review
CBD and THC in Special Populations: Pharmacokinetics and Drug–Drug Interactions
by Lixuan Qian, Jessica L. Beers, Klarissa D. Jackson and Zhu Zhou
Pharmaceutics 2024, 16(4), 484; https://doi.org/10.3390/pharmaceutics16040484 - 1 Apr 2024
Cited by 9 | Viewed by 5892
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 [...] Read more.
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. Full article
(This article belongs to the Special Issue Pharmacokinetics, Pharmacodynamics and Drug Interactions)
11 pages, 1574 KiB  
Article
Pharmacokinetic Interaction of Kratom and Cannabidiol in Male Rats
by Erin C. Berthold, Shyam H. Kamble, Siva Rama Raju Kanumuri, Michelle A. Kuntz, Alexandria S. Senetra, Yi-Hua Chiang, Sushobhan Mukhopadhyay, Christopher R. McCurdy and Abhisheak Sharma
Pharmaceutics 2024, 16(3), 318; https://doi.org/10.3390/pharmaceutics16030318 - 24 Feb 2024
Cited by 4 | Viewed by 4733
Abstract
Kratom and cannabidiol products are used to self-treat a variety of conditions, including anxiety and pain, and to elevate mood. Research into the individual pharmacokinetic properties of commercially available kratom and cannabidiol products has been performed, but there are no studies on coadministration [...] Read more.
Kratom and cannabidiol products are used to self-treat a variety of conditions, including anxiety and pain, and to elevate mood. Research into the individual pharmacokinetic properties of commercially available kratom and cannabidiol products has been performed, but there are no studies on coadministration of these products. Surveys of individuals with kratom use history indicate that cannabidiol use is one of the strongest predictors of both lifetime and past month kratom use. The purpose of this study was to determine if there are changes in pharmacokinetic properties when commercially available kratom and cannabidiol products are administered concomitantly. It was found that with concomitant administration of cannabidiol, there was a 2.8-fold increase in the exposure of the most abundant kratom alkaloid, mitragynine, and increases in the exposure of other minor alkaloids. The results of this work suggest that with cannabidiol coadministration, the effects of kratom may be both delayed and increased due to a delay in time to reach maximum plasma concentration and higher systemic exposure of the psychoactive alkaloids found in kratom. Full article
(This article belongs to the Special Issue Pharmacokinetics, Pharmacodynamics and Drug Interactions)
Show Figures

Figure 1

Back to TopTop