Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 579
Filtrar
1.
Recent Adv Drug Deliv Formul ; 18(4): 247-261, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39356097

RESUMEN

Medical cannabis has potential therapeutic benefits in managing pain, anxiety, depression, and neurological and movement disorders. Phytocannabinoids derived from the cannabis plant are responsible for their pharmacological and therapeutic properties. However, the complexity of cannabis components, especially cannabinoids, poses a challenge to effective medicinal administration. Even with the increasing acceptance of cannabis-based medicines, achieving consistent bioavailability and targeted distribution remains difficult. Conventional administration methods are plagued by solubility and absorption problems requiring innovative solutions. After conducting a thorough review of research papers and patents, it has become evident that nanotechnology holds great promise as a solution. The comprehensive review of 36 research papers has yielded valuable insights, with 7 papers reporting enhanced bioavailability, while others have focused on improvements in release, solubility, and stability. Additionally, 19 patents have been analyzed, of which 7 specifically claim enhanced bioavailability, while the remaining patents describe various formulation methods. These patents outline effective techniques for encapsulating cannabis using nanocarriers, effectively addressing solubility and controlled release. Studies on the delivery of cannabis using nanocarriers focus on improving bioavailability, prolonging release, and targeting specific areas. This synthesis highlights the potential of nanotechnology to enhance cannabis therapies and pave the way for innovative interventions and precision medicine.


Asunto(s)
Cannabinoides , Portadores de Fármacos , Nanopartículas , Humanos , Cannabinoides/química , Cannabinoides/administración & dosificación , Cannabinoides/uso terapéutico , Cannabinoides/farmacología , Cannabinoides/farmacocinética , Portadores de Fármacos/química , Nanopartículas/química , Nanopartículas/uso terapéutico , Disponibilidad Biológica , Marihuana Medicinal/uso terapéutico , Marihuana Medicinal/administración & dosificación , Marihuana Medicinal/química , Marihuana Medicinal/farmacocinética , Animales , Sistemas de Liberación de Medicamentos/métodos , Solubilidad , Nanotecnología/métodos , Patentes como Asunto
3.
Arch Toxicol ; 98(10): 3289-3298, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38955864

RESUMEN

Many fatal intoxications have been reported in connection with the consumption of newer, highly potent synthetic cannabinoids. Yet, a possible postmortem redistribution (PMR) might complicate reliable interpretation of analytical results. Thus, it is necessary to investigate the PMR-potential of new synthetic cannabinoids. The pig model has already proven to be suitable for this purpose. Hence, the aim of this study was to study the PMR of the synthetic cannabinoid 5F-MDMB-P7AICA and its main metabolite 5F-MDMB-P7AICA-dimethylbutanoic acid (DBA). 5F-MDMB-P7AICA (200 µg/kg body weight) was administered by inhalation to anesthetized and ventilated pigs. At the end of the experiment, the animals were euthanized and stored at room temperature for 3 days. Tissue and body fluid samples were taken daily. Specimens were analyzed after solid phase extraction using a standard addition method and LC-MS/MS, blood was quantified after protein precipitation using a validated method. In perimortem samples, 5F-MDMB-P7AICA was found mainly in adipose tissue, bile fluid, and duodenum contents. Small amounts of 5F-MDMB-P7AICA were found in blood, muscle, brain, liver, and lung. High concentrations of DBA were found primarily in bile fluid, duodenum contents, urine, and kidney/perirenal fat tissue. In the remaining tissues, rather low amounts could be found. In comparison to older synthetic cannabinoids, PMR of 5F-MDMB-P7AICA was less pronounced. Concentrations in blood also appear to remain relatively stable at a low level postmortem. Muscle, kidney, fat, and duodenum content are suitable alternative matrices for the detection of 5F-MDMB-P7AICA and DBA, if blood specimens are not available. In conclusion, concentrations of 5F-MDMB-P7AICA and its main metabolite DBA are not relevantly affected by PMR.


Asunto(s)
Líquidos Corporales , Cannabinoides , Cambios Post Mortem , Animales , Cannabinoides/farmacocinética , Cannabinoides/administración & dosificación , Porcinos , Distribución Tisular , Líquidos Corporales/química , Líquidos Corporales/metabolismo , Administración por Inhalación , Espectrometría de Masas en Tándem , Masculino , Indoles/farmacocinética , Indoles/administración & dosificación , Indoles/sangre , Bilis/metabolismo , Bilis/química , Femenino , Tejido Adiposo/metabolismo , Cromatografía Liquida , Pulmón/metabolismo , Pulmón/efectos de los fármacos
4.
Basic Clin Pharmacol Toxicol ; 135(3): 334-344, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39044312

RESUMEN

Tea is a recommended way of administration of prescribed cannabis plant products in Denmark. We aimed to investigate the cannabinoid and terpene doses contained in different teas. We analysed tetrahydrocannabinol (THC), tetrahydrocannabinolic acid (THCA), cannabidiol (CBD), cannabidiolic acid (CBDA), and terpene concentrations in three repeated preparations of each type of tea, and in plant material. In standard tea, concentrations of THC were [median (min-max)] 9.5 (2.3-15), 19 (13-34), and 36 (26-57) µg/mL for products with a labelled content of 6.3%, 14%, and 22% total THC (THC + THCA), respectively. The CBD concentration in tea from a product labelled with 8% total CBD (CBD + CBDA) was 7.5 (1.9-10) µg/mL. Based on this, the recommended starting amount of 0.2 L of the different teas would contain between 0.46 and 11.3 mg THC, and 0.38 to 2.0 mg CBD. Adding creamer before, but not after boiling, increased the THC and CBD concentration 2.3-4.4 and 2.1-fold, respectively. Terpenes were detected in plant material, but not in tea. The study elucidates THC and CBD doses in different teas, which may assist the clinician's choice of cannabis product. Moreover, it underscores the need for caution as administration as tea can result in exposure to different doses, even when the same cannabis product is used.


Asunto(s)
Cannabinoides , Marihuana Medicinal , Terpenos , Marihuana Medicinal/administración & dosificación , Marihuana Medicinal/química , Cannabinoides/análisis , Cannabinoides/administración & dosificación , Cannabinoides/química , Terpenos/análisis , Terpenos/administración & dosificación , Dronabinol/análisis , Dronabinol/administración & dosificación , Cannabidiol/análisis , Cannabidiol/administración & dosificación , Dinamarca , Cannabis/química , Tés Medicinales/análisis
5.
Food Chem Toxicol ; 191: 114848, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38971552

RESUMEN

Spent hemp biomass (SHB) contains trace amounts of cannabinoids, including Δ9-tetrahydrocannabinol (Δ9-THC), that may accumulate in the tissues of animals consuming SHB. We measured cannabinoid residues in the liver, adipose tissue, and muscle of finishing lambs fed either 10% or 20% SHB for 8 weeks, or 4 weeks followed by 4 weeks SHB withdrawal. We detected multiple cannabinoids in the liver at a similar proportion to the SHB. However, CBD and Δ9-THC were enriched >20-fold in the adipose and muscle, compared to their proportion in SHB. The highest concentration of Δ9-THC was detected in adipose tissue and was 7.4-times higher than in muscle. Most cannabinoids were undetectable in tissues after 4 weeks of clearance. The consumers' exposure assessment on Δ9-THC revealed tissue levels of total THC (THCA+Δ9-THC) that exceed the acute reference dose of 1 µg/kg BW across population groups. When consuming meat from the lambs fed 10% and 20% SHB, the maximum total THC exposure was 2.03 and 7.32 µg/kg BW, respectively, equal to or below the Lowest Observed Adverse Effect Level of 36 µg/kg BW, the No Observed Adverse Effect Level of 12 µg/kg BW or a tolerable dose intake of 7 µg/kg BW.


Asunto(s)
Cannabinoides , Cannabis , Animales , Cannabis/química , Cannabinoides/análisis , Cannabinoides/administración & dosificación , Ovinos , Alimentación Animal/análisis , Hígado/metabolismo , Hígado/química , Hígado/efectos de los fármacos , Contaminación de Alimentos/análisis , Humanos , Biomasa , Tejido Adiposo/metabolismo , Tejido Adiposo/química , Dronabinol/análisis
6.
J Nutr Health Aging ; 28(8): 100299, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38917597

RESUMEN

BACKGROUND AND OBJECTIVES: Anorexia of aging (AoA) is a prevalent geriatric syndrome characterized by a multifactorial decline in appetite and reduced food intake associated with the aging process. This systematic review aims to investigate the use and outcomes of cannabinoids in addressing AoA, with the goal of providing a comprehensive understanding and discussing their potential integration into daily clinical practice. METHODS: A thorough search of databases (Embase Ovid, Scopus, PubMed, Cochrane Library, and Web of Science) identified 6100 studies. After eliminating duplicates and screening titles and abstracts, 25 studies underwent full appraisal. Two reviewers assessed inclusion suitability, and study methodologies were evaluated using the Newcastle-Ottawa Scale (NOS) for observational studies and the modified Jadad Scoring Scale for randomized controlled trials. Ultimately, six studies published between 2002 and 2019, involving 869 participants, were included in the review. RESULTS: Out of the 6 fin. l papers selected, 5 were randomized trials, and 1 was a prospective study. Megestrol acetate (800 mg/d) proved to be more effective than dronabinol 2.5 mg twice a day in increasing appetite. Nabilone (at a dosage of 0.5 mg per day) did not show superiority over placebo in alleviating symptoms such as pain, nausea, loss of appetite, and weight. However, with a double dosage followed by 1.0 mg/6 weeks, after eight weeks of treatment, patients recorded a significant increase in calorie intake and carbohydrate consumption compared to the placebo group, with some patients also experiencing substantial weight gain. Regarding delta-9-tetrahydrocannabinol (THC), a weight increase of ≥10% was observed in 17.6% of patients with doses of 5 mg or 10 mg capsules daily, without significant side effects. Additionally, patients treated with THC 2.5 mg reported improved chemosensory perception and increased appetite before meals compared to placebo. No significant side effects were reported in older adults taking cannabinoids. CONCLUSIONS: Cannabinoids offer promise in enhancing the quality of life for older individuals with active neoplastic disease. However, to establish comprehensive guidelines, further research with larger sample sizes is essential. Only through this approach can we fully grasp the potential and application of cannabinoids in addressing the nutritional concerns associated with neoplastic diseases.


Asunto(s)
Anorexia , Cannabinoides , Neoplasias , Humanos , Anorexia/tratamiento farmacológico , Cannabinoides/administración & dosificación , Cannabinoides/uso terapéutico , Anciano , Neoplasias/complicaciones , Neoplasias/tratamiento farmacológico , Apetito/efectos de los fármacos , Dronabinol/análogos & derivados , Femenino , Masculino , Ensayos Clínicos Controlados Aleatorios como Asunto , Acetato de Megestrol/uso terapéutico , Acetato de Megestrol/administración & dosificación
7.
J Psychiatr Res ; 175: 50-59, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38704981

RESUMEN

Major depressive disorder (MDD) stands as a significant cause of disability globally. Cannabidiolic Acid-Methyl Ester (CBDA-ME) (EPM-301, HU-580), a derivative of Cannabidiol, demonstrates immediate antidepressant-like effects, yet it has undergone only minimal evaluation in psychopharmacology. Our goal was to investigate the behavioral and potential molecular mechanisms associated with the chronic oral administration of this compound in the Wistar Kyoto (WKY) genetic model of treatment-resistant depression. Male WKY rats were subjected to behavioral assessments before and after receiving chronic (14-day) oral doses of CBDA-ME (0.5 mg/kg), 15 mg/kg of imipramine or vehicle. At the end of the study, plasma corticosterone levels and mRNA expression of various genes in the medial Prefrontal Cortex and Hippocampus were measured. Behavioral outcomes from CBDA-ME treatment indicated an antidepressant-like effect similar to imipramine, as oral ingestion reduced immobility and increased swimming duration in the Forced Swim Test. Neither treatment influenced locomotion in the Open Field Test nor preference in the Saccharin Preference Test. The behavioral impact in WKY rats coincided with reduced corticosterone serum levels, upregulated mRNA expression of Cannabinoid receptor 1, Fatty Acid Amide Hydrolase, and Corticotropin-Releasing Hormone Receptor 1, alongside downregulation of the Serotonin Transporter in the hippocampus. Additionally, there was an upregulation of CB1 mRNA expression and downregulation of Brain-Derived Neurotrophic Factor in the mPFC. These findings contribute to our limited understanding of the antidepressant effects of CBDA-ME and shed light on its potential psychopharmacological mechanisms. This discovery opens up possibilities for utilizing cannabinoids in the treatment of major depressive disorder and related conditions.


Asunto(s)
Antidepresivos , Trastorno Depresivo Resistente al Tratamiento , Modelos Animales de Enfermedad , Imipramina , Corteza Prefrontal , Ratas Endogámicas WKY , Animales , Masculino , Ratas , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Antidepresivos/farmacología , Antidepresivos/administración & dosificación , Imipramina/farmacología , Imipramina/administración & dosificación , Trastorno Depresivo Resistente al Tratamiento/tratamiento farmacológico , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Cannabinoides/farmacología , Cannabinoides/administración & dosificación , Corticosterona/sangre , Conducta Animal/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/efectos de los fármacos , Receptor Cannabinoide CB1/metabolismo , Receptor Cannabinoide CB1/efectos de los fármacos , Amidohidrolasas/metabolismo
8.
Int J Nanomedicine ; 19: 4607-4649, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38799700

RESUMEN

Cannabinoids are compounds found in and derived from the Cannabis plants that have become increasingly recognised as significant modulating factors of physiological mechanisms and inflammatory reactions of the organism, thus inevitably affecting maintenance of homeostasis. Medical Cannabis popularity has surged since its legal regulation growing around the world. Numerous promising discoveries bring more data on cannabinoids' pharmacological characteristics and therapeutic applications. Given the current surge in interest in the medical use of cannabinoids, there is an urgent need for an effective method of their administration. Surpassing low bioavailability, low water solubility, and instability became an important milestone in the advancement of cannabinoids in pharmaceutical applications. The numerous uses of cannabinoids in clinical practice remain restricted by limited administration alternatives, but there is hope when biodegradable polymers are taken into account. The primary objective of this review is to highlight the wide range of indications for which cannabinoids may be used, as well as the polymeric carriers that enhance their effectiveness. The current review described a wide range of therapeutic applications of cannabinoids, including pain management, neurological and sleep disorders, anxiety, and cancer treatment. The use of these compounds was further examined in the area of dermatology and cosmetology. Finally, with the use of biodegradable polymer-based drug delivery systems (DDSs), it was demonstrated that cannabinoids can be delivered specifically to the intended site while also improving the drug's physicochemical properties, emphasizing their utility. Nevertheless, additional clinical trials on novel cannabinoids' formulations are required, as their full spectrum therapeutical potential is yet to be unravelled.


Asunto(s)
Cannabinoides , Polímeros , Humanos , Cannabinoides/química , Cannabinoides/administración & dosificación , Cannabinoides/farmacocinética , Cannabinoides/farmacología , Polímeros/química , Sistemas de Liberación de Medicamentos/métodos , Animales , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Manejo del Dolor/métodos
9.
Int J Mol Sci ; 25(8)2024 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-38673862

RESUMEN

Chemotherapy-induced peripheral neuropathy (CIPN) is one of the most prevalent and dose-limiting complications in chemotherapy patients. One identified mechanism underlying CIPN is neuroinflammation. Most of this research has been conducted in only male or female rodent models, making direct comparisons regarding the role of sex differences in the neuroimmune underpinnings of CIPN limited. Moreover, most measurements have focused on the dorsal root ganglia (DRG) and/or spinal cord, while relatively few studies have been aimed at characterizing neuroinflammation in the brain, for example the periaqueductal grey (PAG). The overall goals of the present study were to determine (1) paclitaxel-associated changes in markers of inflammation in the PAG and DRG in male and female C57Bl6 mice and (2) determine the effect of prophylactic administration of an anti-inflammatory cannabinoid, cannabigerol (CBG). In Experiment 1, male and female mice were treated with paclitaxel (8-32 mg/kg/injection, Days 1, 3, 5, and 7) and mechanical sensitivity was measured using Von Frey filaments on Day 7 (Cohort 1) and Day 14 (Cohort 2). Cohorts were euthanized on Day 8 or 15, respectively, and DRG and PAG were harvested for qPCR analysis of the gene expression of markers of pain and inflammation Aig1, Gfap, Ccl2, Cxcl9, Tlr4, Il6, and Calca. In Experiment 2, male and female mice were treated with vehicle or 10 mg/kg CBG i.p. 30 min prior to each paclitaxel injection. Mechanical sensitivity was measured on Day 14. Mice were euthanized on Day 15, and PAG were harvested for qPCR analysis of the gene expression of Aig1, Gfap, Ccl2, Cxcl9, Tlr4, Il6, and Calca. Paclitaxel produced a transient increase in potency to produce mechanical sensitivity in male versus female mice. Regarding neuroinflammation, more gene expression changes were apparent earlier in the DRG and at a later time point in the PAG. Also, more changes were observed in females in the PAG than males. Overall, sex differences were observed for most markers at both time points and regions. Importantly, in both the DRG and PAG, most increases in markers of neuroinflammation and pain occurred at paclitaxel doses higher than those associated with significant changes in the mechanical threshold. Two analytes that demonstrated the most compelling sexual dimorphism and that changed more in males were Cxcl9 and Ccl2, and Tlr4 in females. Lastly, prophylactic administration of CBG protected the male and female mice from increased mechanical sensitivity and female mice from neuroinflammation in the PAG. Future studies are warranted to explore how these sex differences may shed light on the mechanisms of CIPN and how non-psychoactive cannabinoids such as CBG may engage these targets to prevent or attenuate the effects of paclitaxel and other chemotherapeutic agents on the nervous system.


Asunto(s)
Ratones Endogámicos C57BL , Paclitaxel , Animales , Paclitaxel/efectos adversos , Femenino , Masculino , Ratones , Cannabinoides/farmacología , Cannabinoides/administración & dosificación , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Ganglios Espinales/metabolismo , Ganglios Espinales/efectos de los fármacos , Factores Sexuales , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/inducido químicamente , Hiperalgesia/metabolismo , Caracteres Sexuales , Inflamación/tratamiento farmacológico , Inflamación/metabolismo
10.
Curr Pharm Des ; 30(12): 921-934, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38482628

RESUMEN

BACKGROUND: Transdermal delivery of highly lipophilic molecules is challenging due to the strong barrier function of the skin. Vesicles with penetration enhancers are safe and efficient systems that could improve the transdermal delivery of non-psychoactive cannabinoids such as cannabidiol and desoxy-cannabidiol. In the last decades, research interest in desoxy-cannabidiol as a potent drug with anti-nociceptive properties has risen. Still, its scarce market availability poses a limit for both research and clinical applications. Therefore, it is necessary to improve the synthesis to produce sufficient amounts of desoxy-cannabidiol. Moreover, also the formulation aspects for this drug are challenging and require to be addressed to meet an efficient delivery to the patients. OBJECTIVE: This work aimed to develop innovative phospholipid-based vesicles with propylene glycol (PG), oleic acid (OA), or limonene as edge activators, for the transdermal delivery of highly lipophilic drugs such as non-psychoactive cannabinoids. In particular, desoxy-cannabidiol was selected thanks to its anti-nociceptive activity, and its synthesis was improved enhancing the stereoselectivity of its synthon's production. METHODS: Desoxy-cannabidiol was synthesized by Lewis acid-mediated condensation of p-mentha-2,8-dien- 1-ol and m-pentylphenol, improving the stereoselectivity of the first synthon's production. Transethosomes containing 20-50% w/w PG, 0.4-0.8% w/w OA, or 0.1-1% w/w limonene were optimized and loaded with cannabidiol or desoxy-cannabidiol (0.07-0.8% w/w, 0.6-7.0 mg/mL). Ex-vivo studies were performed to assess both the skin permeation and accumulation of the cannabinoids, as well as the penetration depth of fluorescein- loaded systems used as models. RESULTS: An enantioselective bromination was added to the pathway, thus raising the production yield of pmentha- 2,8-dien-1-ol to 81% against 35%, and the overall yield of desoxy-cannabidiol synthesis from 12% to 48%. Optimized transethosomes containing 0.6 mg/mL cannabinoids were prepared with 1:10 PG:lipid weight ratio, 0.54 OA:lipid molar ratio, and 0.3 limonene:lipid molar ratio, showing good nanometric size (208 ± 20.8 nm - 321 ± 26.3 nm) and entrapment efficiency (> 80%). Ex-vivo tests showed both improved skin permeation rates of cannabinoids (up to 21.32 ± 4.27 µg/cm2 cannabidiol), and skin penetration (depth of fluorescein up to 240 µm, with PG). CONCLUSION: Desoxy-cannabidiol was successfully produced at high yields, and formulated into transethosomes optimized for transdermal delivery. Loaded vesicles showed improved skin penetration of desoxy-cannabidiol, cannabidiol and a lipophilic probe. These results suggest the potential of these carriers for the transdermal delivery of highly lipophilic drugs.


Asunto(s)
Administración Cutánea , Cannabinoides , Sistemas de Liberación de Medicamentos , Absorción Cutánea , Cannabinoides/administración & dosificación , Cannabinoides/química , Cannabinoides/síntesis química , Cannabinoides/farmacocinética , Animales , Absorción Cutánea/efectos de los fármacos , Piel/metabolismo , Piel/efectos de los fármacos , Humanos , Cannabidiol/administración & dosificación , Cannabidiol/farmacocinética , Cannabidiol/química , Ratas , Masculino , Estructura Molecular
11.
J Clin Pharmacol ; 64(5): 499-513, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38145388

RESUMEN

Cannabis-based therapeutics have garnered increasing attention in recent years as patients seek alternative treatments for various medical conditions. This narrative review provides a comprehensive overview of the science behind the medical use of cannabis, focusing on the medical evidence for commonly treated conditions. In addition, the review addresses the practical considerations of using cannabis as a therapeutic agent, offering insights into dosing strategies, variations in cannabinoid formulation, and individual patient responses. Precautions, adverse consequences, and drug interactions are also discussed, with a focus on patient safety and the potential risks associated with cannabis use.


Asunto(s)
Cannabis , Marihuana Medicinal , Humanos , Cannabinoides/uso terapéutico , Cannabinoides/farmacología , Cannabinoides/administración & dosificación , Cannabis/química , Interacciones Farmacológicas , Marihuana Medicinal/uso terapéutico , Marihuana Medicinal/efectos adversos
12.
Adv Ther ; 40(1): 282-293, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36308640

RESUMEN

INTRODUCTION: Transdermal cannabinoids may provide better safety and bioavailability profiles compared with other routes of administration. This single-arm, open-label study investigated a novel topical transdermal delivery system on the pharmacokinetics of cannabidiol (CBD) and tetrahydrocannabinol (THC). METHODS: Participants were 39.5 ± 7.37 years old and healthy, based on a review by the Medical Director. Blood was collected pre-dose and 10, 20, 30, and 45 min, and 1, 1.5, 2, 2.5, 3, 4, 5, 6, 8, and 12 h after topical application of 100 mg CBD:100 mg THC. Psychoactive effects were assessed prior to each timepoint. Area-under-the-curve (AUC0-12 h), maximum concentration (Cmax), time to maximum concentration (Tmax), area-under-the-curve to infinity (AUCI), terminal elimination rate constant (λ), terminal half-life (t½), and absorption rate constant (ka) were measured individually for CBD and THC. Safety was assessed by clinical chemistry, hematology, and adverse events. RESULTS: AUC0-12 h for CBD and THC was 3329.8 ± 3252.1 and 2093.4 ± 2090.6 pg/mL/h, with Cmax of 576.52 ± 1016.18 and 346.57 ± 776.85 pg/mL, respectively. Tmax for CBD and THC was 8 h, ranging from 2.5 h to 12 h and 10 min to 12 h, respectively. AUCI for CBD and THC was 6609.2 ± 7056.4 and 3721.0 ± 3251.7 pg/mL/h, with t1/2 of 5.68 ± 1.5 and 5.38 ± 1.25 h, respectively. CBD was absorbed at a faster rate compared with THC (123.36 ± 530.97 versus 71.5 ± 1142.19 h-1) but with similar λ (0.12 ± 0.029 versus 0.13 ± 0.03 h-1). No psychoactive effects were reported. Transdermal cannabinoid delivery was safe and well tolerated in the population studied. CONCLUSION: To our knowledge, this is the first pharmacokinetic study in humans that demonstrated CBD and THC entering systemic circulation via transdermal administration . This study represents an important contribution to understanding the pharmacokinetics of transdermal cannabinoids. CLINICAL TRIAL REGISTRATION: ClinicalTrials.gov Identifier-NCT05121506 (November 16, 2021).


Asunto(s)
Cannabidiol , Dronabinol , Adulto , Humanos , Persona de Mediana Edad , Administración Cutánea , Disponibilidad Biológica , Cannabidiol/administración & dosificación , Cannabidiol/farmacocinética , Cannabinoides/administración & dosificación , Cannabinoides/efectos adversos , Dronabinol/administración & dosificación , Dronabinol/farmacocinética
13.
PLoS One ; 17(1): e0262633, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35045097

RESUMEN

Currently, peripheral tissue distribution of cannabinoids after treatment is poorly understood. This pilot study sought to examine the early tissue distribution of major cannabinoids 30 minutes following an intraperitoneal injection of vehicle (1:9 Tween 80/SAL), and doses of THC (1 mg/kg) and CBD (5 mg/kg) that are feasible for human consumption in serum, adipose, brain, lung, liver, jejunum, and muscle of male Sprague-Dawley rats. The jejunum and adipose were most enriched in THC. Similarly, CBD was enriched in the jejunum and adipose but also the liver. In contrast, the brain had the lowest concentration of cannabinoids relative to other tissues. The liver had the greatest concentration of the THC metabolites, 11-OH-THC and COOH-THC, compared to all other tissues. Overall, these findings highlight broad tissue distribution and marked differences in tissue concentration not previously appreciated. Thus, as cannabinoid research continues to rapidly grow, consideration of the potential bioactive effects of these molecules in peripheral tissues is warranted in future studies.


Asunto(s)
Cannabinoides/administración & dosificación , Cannabinoides/farmacología , Distribución Tisular/fisiología , Animales , Cannabinoides/metabolismo , Inyecciones Intraperitoneales , Masculino , Proyectos Piloto , Ratas , Ratas Sprague-Dawley , Distribución Tisular/efectos de los fármacos
15.
Sci Rep ; 11(1): 22776, 2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34815467

RESUMEN

Legalization of cannabis for medicinal and/or recreational use is expanding globally. Although cannabis is being regulated country by country, an accurate recent use test with indisputable results correlated with impairment has yet to be discovered. In the present study, a new approach for determining recent cannabis use within the impairment window after smoking was developed by studying 74 subjects with a mean age of 25 years and average use history of 9 years. Horizontal gaze nystagmus was evaluated along with subject self-assessments of impairment, and blood and breath samples were collected before and after smoking cannabis. Breath and blood pharmacokinetic parameters and cannabinoid profiles determined recent use within the impairment window. No subjects were positive for recent use pre-smoking, although all subjects had detectable cannabinoids in breath samples. We describe an inhaled cannabis recent use test that correlates with impairment and helps protect against wrongful prosecution and workplace discrimination.


Asunto(s)
Pruebas Respiratorias/métodos , Cannabinoides/análisis , Cannabis/química , Detección de Abuso de Sustancias/métodos , Administración por Inhalación , Adulto , Cannabinoides/administración & dosificación , Femenino , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
16.
Eur J Pharmacol ; 911: 174560, 2021 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-34648805

RESUMEN

The global incidence of respiratory diseases and complications is increasing. Therefore, new methods of treatment, as well as prevention, need to be investigated. A group of compounds that should be considered for use in respiratory diseases is cannabinoids. There are three groups of cannabinoids - plant-derived phytocannabinoids, synthetic cannabinoids, and endogenous endocannabinoids including the enzymes responsible for their synthesis and degradation. All cannabinoids exert their biological effects through either type 1 cannabinoid receptors (CB1) and/or type 2 cannabinoid receptors (CB2). In numerous studies (in vitro and in vivo), cannabinoids and inhibitors of endocannabinoid degradation have shown beneficial anti-inflammatory, antioxidant, anti-cancer, and anti-fibrotic properties. Although in the respiratory system, most of the studies have focused on the positive properties of cannabinoids and inhibitors of endocannabinoid degradation. There are few research reports discussing the negative impact of these compounds. This review summarizes the properties and mechanisms of action of cannabinoids and inhibitors of endocannabinoid degradation in various models of respiratory diseases. A short description of the effects selected cannabinoids have on the human respiratory system and their possible use in the fight against COVID-19 is also presented. Additionally, a brief summary is provided of cannabinoid receptors properties and their expression in the respiratory system and cells of the immune system.


Asunto(s)
Cannabinoides/farmacología , Endocannabinoides/metabolismo , Enfermedades Respiratorias/tratamiento farmacológico , Animales , Cannabinoides/administración & dosificación , Inhibidores Enzimáticos/farmacología , Humanos , Modelos Biológicos , Receptores de Cannabinoides/inmunología , Receptores de Cannabinoides/metabolismo , Enfermedades Respiratorias/metabolismo , Tratamiento Farmacológico de COVID-19
17.
BMJ ; 374: n1034, 2021 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-34497047

RESUMEN

OBJECTIVE: To determine the benefits and harms of medical cannabis and cannabinoids for chronic pain. DESIGN: Systematic review and meta-analysis. DATA SOURCES: MEDLINE, EMBASE, AMED, PsycInfo, CENTRAL, CINAHL, PubMed, Web of Science, Cannabis-Med, Epistemonikos, and trial registries up to January 2021. STUDY SELECTION: Randomised clinical trials of medical cannabis or cannabinoids versus any non-cannabis control for chronic pain at ≥1 month follow-up. DATA EXTRACTION AND SYNTHESIS: Paired reviewers independently assessed risk of bias and extracted data. We performed random-effects models meta-analyses and used GRADE to assess the certainty of evidence. RESULTS: A total of 32 trials with 5174 adult patients were included, 29 of which compared medical cannabis or cannabinoids with placebo. Medical cannabis was administered orally (n=30) or topically (n=2). Clinical populations included chronic non-cancer pain (n=28) and cancer related pain (n=4). Length of follow-up ranged from 1 to 5.5 months. Compared with placebo, non-inhaled medical cannabis probably results in a small increase in the proportion of patients experiencing at least the minimally important difference (MID) of 1 cm (on a 10 cm visual analogue scale (VAS)) in pain relief (modelled risk difference (RD) of 10% (95% confidence interval 5% to 15%), based on a weighted mean difference (WMD) of -0.50 cm (95% CI -0.75 to -0.25 cm, moderate certainty)). Medical cannabis taken orally results in a very small improvement in physical functioning (4% modelled RD (0.1% to 8%) for achieving at least the MID of 10 points on the 100-point SF-36 physical functioning scale, WMD of 1.67 points (0.03 to 3.31, high certainty)), and a small improvement in sleep quality (6% modelled RD (2% to 9%) for achieving at least the MID of 1 cm on a 10 cm VAS, WMD of -0.35 cm (-0.55 to -0.14 cm, high certainty)). Medical cannabis taken orally does not improve emotional, role, or social functioning (high certainty). Moderate certainty evidence shows that medical cannabis taken orally probably results in a small increased risk of transient cognitive impairment (RD 2% (0.1% to 6%)), vomiting (RD 3% (0.4% to 6%)), drowsiness (RD 5% (2% to 8%)), impaired attention (RD 3% (1% to 8%)), and nausea (RD 5% (2% to 8%)), but not diarrhoea; while high certainty evidence shows greater increased risk of dizziness (RD 9% (5% to 14%)) for trials with <3 months follow-up versus RD 28% (18% to 43%) for trials with ≥3 months follow-up; interaction test P=0.003; moderate credibility of subgroup effect). CONCLUSIONS: Moderate to high certainty evidence shows that non-inhaled medical cannabis or cannabinoids results in a small to very small improvement in pain relief, physical functioning, and sleep quality among patients with chronic pain, along with several transient adverse side effects, compared with placebo. The accompanying BMJ Rapid Recommendation provides contextualised guidance based on this body of evidence. SYSTEMATIC REVIEW REGISTRATION: https://osf.io/3pwn2.


Asunto(s)
Dolor en Cáncer/tratamiento farmacológico , Cannabinoides/efectos adversos , Dolor Crónico/tratamiento farmacológico , Marihuana Medicinal/administración & dosificación , Adulto , Cannabinoides/administración & dosificación , Femenino , Humanos , Masculino , Marihuana Medicinal/efectos adversos , Diferencia Mínima Clínicamente Importante , Dimensión del Dolor , Ensayos Clínicos Controlados Aleatorios como Asunto , Sueño/efectos de los fármacos
18.
BMJ ; 374: n2040, 2021 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-34497062

RESUMEN

CLINICAL QUESTION: What is the role of medical cannabis or cannabinoids for people living with chronic pain due to cancer or non-cancer causes? CURRENT PRACTICE: Chronic pain is common and distressing and associated with considerable socioeconomic burden globally. Medical cannabis is increasingly used to manage chronic pain, particularly in jurisdictions that have enacted policies to reduce use of opioids; however, existing guideline recommendations are inconsistent, and cannabis remains illegal for therapeutic use in many countries. RECOMMENDATION: The guideline expert panel issued a weak recommendation to offer a trial of non-inhaled medical cannabis or cannabinoids, in addition to standard care and management (if not sufficient), for people living with chronic cancer or non-cancer pain. HOW THIS GUIDELINE WAS CREATED: An international guideline development panel including patients, clinicians with content expertise, and methodologists produced this recommendation in adherence with standards for trustworthy guidelines using the GRADE approach. The MAGIC Evidence Ecosystem Foundation (MAGIC) provided methodological support. The panel applied an individual patient perspective. THE EVIDENCE: This recommendation is informed by a linked series of four systematic reviews summarising the current body of evidence for benefits and harms, as well as patient values and preferences, regarding medical cannabis or cannabinoids for chronic pain. UNDERSTANDING THE RECOMMENDATION: The recommendation is weak because of the close balance between benefits and harms of medical cannabis for chronic pain. It reflects a high value placed on small to very small improvements in self reported pain intensity, physical functioning, and sleep quality, and willingness to accept a small to modest risk of mostly self limited and transient harms. Shared decision making is required to ensure patients make choices that reflect their values and personal context. Further research is warranted and may alter this recommendation.


Asunto(s)
Cannabinoides/administración & dosificación , Dolor Crónico/tratamiento farmacológico , Marihuana Medicinal/administración & dosificación , Adolescente , Adulto , Cannabinoides/efectos adversos , Niño , Humanos , Marihuana Medicinal/efectos adversos , Adulto Joven
19.
Front Neural Circuits ; 15: 662349, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34305536

RESUMEN

Our sensory systems such as the olfactory and visual systems are the target of neuromodulatory regulation. This neuromodulation starts at the level of sensory receptors and extends into cortical processing. A relatively new group of neuromodulators includes cannabinoids. These form a group of chemical substances that are found in the cannabis plant. Δ9-tetrahydrocannabinol (THC) and cannabidiol (CBD) are the main cannabinoids. THC acts in the brain and nervous system like the chemical substances that our body produces, the endogenous cannabinoids or endocannabinoids, also nicknamed the brain's own cannabis. While the function of the endocannabinoid system is understood fairly well in limbic structures such as the hippocampus and the amygdala, this signaling system is less well understood in the olfactory pathway and the visual system. Here, we describe and compare endocannabinoids as signaling molecules in the early processing centers of the olfactory and visual system, the olfactory bulb, and the retina, and the relevance of the endocannabinoid system for synaptic plasticity.


Asunto(s)
Cannabinoides/metabolismo , Plasticidad Neuronal/fisiología , Bulbo Olfatorio/metabolismo , Olfato/fisiología , Vías Visuales/metabolismo , Percepción Visual/fisiología , Animales , Cannabinoides/administración & dosificación , Humanos , Plasticidad Neuronal/efectos de los fármacos , Bulbo Olfatorio/efectos de los fármacos , Receptor Cannabinoide CB1/agonistas , Receptor Cannabinoide CB1/metabolismo , Retina/efectos de los fármacos , Retina/metabolismo , Olfato/efectos de los fármacos , Vías Visuales/efectos de los fármacos , Percepción Visual/efectos de los fármacos
20.
Sci Rep ; 11(1): 14948, 2021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34294753

RESUMEN

Cannabis is a complex mixture of hundreds of bioactive molecules. This provides the potential for pharmacological interactions between cannabis constituents, a phenomenon referred to as "the entourage effect" by the medicinal cannabis community. We hypothesize that pharmacokinetic interactions between cannabis constituents could substantially alter systemic cannabinoid concentrations. To address this hypothesis we compared pharmacokinetic parameters of cannabinoids administered orally in a cannabis extract to those administered as individual cannabinoids at equivalent doses in mice. Astonishingly, plasma cannabidiolic acid (CBDA) concentrations were 14-times higher following administration in the cannabis extract than when administered as a single molecule. In vitro transwell assays identified CBDA as a substrate of the drug efflux transporter breast cancer resistance protein (BCRP), and that cannabigerol and Δ9-tetrahydrocannabinol inhibited the BCRP-mediated transport of CBDA. Such a cannabinoid-cannabinoid interaction at BCRP transporters located in the intestine would inhibit efflux of CBDA, thus resulting in increased plasma concentrations. Our results suggest that cannabis extracts provide a natural vehicle to substantially enhance plasma CBDA concentrations. Moreover, CBDA might have a more significant contribution to the pharmacological effects of orally administered cannabis extracts than previously thought.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Cannabinoides/administración & dosificación , Cannabis/química , Aceites de Plantas/administración & dosificación , Administración Oral , Animales , Disponibilidad Biológica , Cannabinoides/sangre , Cannabinoides/química , Cannabinoides/farmacocinética , Suplementos Dietéticos , Perros , Células de Riñón Canino Madin Darby , Ratones , Modelos Animales , Aceites de Plantas/química , Aceites de Plantas/farmacocinética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA