Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 199
Filter
Add more filters

Complementary Medicines
Publication year range
1.
Sci Rep ; 12(1): 2449, 2022 02 14.
Article in English | MEDLINE | ID: mdl-35165360

ABSTRACT

Resting state fMRI has been employed to identify alterations in functional connectivity within or between brain regions following acute and chronic exposure to Δ9-tetrahydrocannabinol (THC), the psychoactive component in cannabis. Most studies focused a priori on a limited number of local brain areas or circuits, without considering the impact of cannabis on whole-brain network organization. The present study attempted to identify changes in the whole-brain human functional connectome as assessed with ultra-high field (7T) resting state scans of cannabis users (N = 26) during placebo and following vaporization of cannabis. Two distinct data-driven methodologies, i.e. network-based statistics (NBS) and connICA, were used to identify changes in functional connectomes associated with acute cannabis intoxication and history of cannabis use. Both methodologies revealed a broad state of hyperconnectivity within the entire range of major brain networks in chronic cannabis users compared to occasional cannabis users, which might be reflective of an adaptive network reorganization following prolonged cannabis exposure. The connICA methodology also extracted a distinct spatial connectivity pattern of hypoconnectivity involving the dorsal attention, limbic, subcortical and cerebellum networks and of hyperconnectivity between the default mode and ventral attention network, that was associated with the feeling of subjective high during THC intoxication. Whole-brain network approaches identified spatial patterns in functional brain connectomes that distinguished acute from chronic cannabis use, and offer an important utility for probing the interplay between short and long-term alterations in functional brain dynamics when progressing from occasional to chronic use of cannabis.


Subject(s)
Brain/diagnostic imaging , Brain/physiopathology , Cannabis/chemistry , Connectome/methods , Dronabinol/administration & dosage , Marijuana Smoking/physiopathology , Marijuana Smoking/psychology , Plant Extracts/administration & dosage , Psychotropic Drugs/administration & dosage , Adult , Attention/drug effects , Cognition/drug effects , Cross-Over Studies , Double-Blind Method , Emotions/drug effects , Female , Humans , Magnetic Resonance Imaging/methods , Male , Young Adult
2.
Int J Mol Sci ; 22(16)2021 Aug 18.
Article in English | MEDLINE | ID: mdl-34445602

ABSTRACT

Cannabis is the most-used recreational drug worldwide, with a high prevalence of use among adolescents. In animal models, long-term adverse effects were reported following chronic adolescent exposure to the main psychotomimetic component of the plant, delta-9-tetrahydrocannabinol (THC). However, these studies investigated the effects of pure THC, without taking into account other cannabinoids present in the cannabis plant. Interestingly, cannabidiol (CBD) content seems to mitigate some of the side effects of THC, at least in adult animals. Thus, in female rats, we evaluated the long-term consequences of a co-administration of THC and CBD at a 3:1 ratio, chosen based on the analysis of recently confiscated illegal cannabis samples in Europe. CBD content is able to mitigate some of the long-term behavioral alterations induced by adolescent THC exposure as well as long-term changes in CB1 receptor and microglia activation in the prefrontal cortex (PFC). We also investigated, for the first time, possible long-term effects of chronic administration of a THC/CBD combination reminiscent of "light cannabis" (CBD:THC in a 33:1 ratio; total THC 0.3%). Repeated administration of this CBD:THC combination has long-term adverse effects on cognition and leads to anhedonia. Concomitantly, it boosts Glutamic Acid Decarboxylase-67 (GAD67) levels in the PFC, suggesting a possible lasting effect on GABAergic neurotransmission.


Subject(s)
Behavior, Animal/drug effects , Cannabidiol/administration & dosage , Cognition/drug effects , Dronabinol/administration & dosage , Hallucinogens/administration & dosage , Prefrontal Cortex/drug effects , Animals , Female , Memory/drug effects , Rats , Rats, Sprague-Dawley , Synaptic Transmission
3.
Pharmacol Biochem Behav ; 207: 173222, 2021 08.
Article in English | MEDLINE | ID: mdl-34197845

ABSTRACT

RATIONALE: Despite a long history of use in synaptic physiology, the lobster has been a neglected model for behavioral pharmacology. A restaurateur proposed that exposing lobster to cannabis smoke reduces anxiety and pain during the cooking process. It is unknown if lobster gill respiration in air would result in significant Δ9-tetrahydrocannabinol (THC) uptake and whether this would have any detectable behavioral effects. OBJECTIVE: The primary goal was to determine tissue THC levels in the lobster after exposure to THC vapor. Secondary goals were to determine if THC vapor altered locomotor behavior or nociception. METHODS: Tissue samples were collected (including muscle, brain and hemolymph) from Homarus americanus (N = 3 per group) following 30 or 60 min of exposure to vapor generated by an e-cigarette device using THC (100 mg/mL in a propylene glycol vehicle). Separate experiments assessed locomotor behavior and hot water nociceptive responses following THC vapor exposure. RESULTS: THC vapor produced duration-related THC levels in all tissues examined. Locomotor activity was decreased (distance, speed, time-mobile) by 30 min inhalation of THC. Lobsters exhibit a temperature-dependent withdrawal response to immersion of tail, antennae or claws in warm water; this is novel evidence of thermal nociception for this species. THC exposure for 60 min had only marginal effect on nociception under the conditions assessed. CONCLUSIONS: Vapor exposure of lobsters, using an e-cigarette based model, produces dose-dependent THC levels in all tissues and reduces locomotor activity. Hot water nociception was temperature dependent, but only minimal anti-nociceptive effect of THC exposure was confirmed.


Subject(s)
Dronabinol/pharmacology , E-Cigarette Vapor/pharmacology , Locomotion/drug effects , Nephropidae , Nociception/drug effects , Administration, Inhalation , Animals , Cooking/methods , Dronabinol/administration & dosage , Dronabinol/analysis , E-Cigarette Vapor/administration & dosage , Electronic Nicotine Delivery Systems , Female , Hot Temperature , Maine , Male , Marijuana Smoking/metabolism , Pain/drug therapy , Rats
4.
Behav Brain Res ; 410: 113342, 2021 07 23.
Article in English | MEDLINE | ID: mdl-33961911

ABSTRACT

Using marijuana has become popular and is allowed for medical purposes in some countries. The effect of marijuana on Parkinson's disease is controversial and Medical marijuana may benefit for motor and non-motor symptoms of patients with Parkinson's disease. No research has been conducted to fully prove the benefits, risks, and uses of marijuana as a treatment for patients with Parkinson's disease. In the present study, several different approaches, including behavioral measures and the western blot method for protein level assay, were used to investigate whether exposure to marijuana affects the motor and synaptic plasticity impairment induced by 6-OHDA. Marijuana consumption significantly decreased apomorphine-induced contralateral rotation, beam travel time, beam freeze time, and catalepsy time, but significantly increased latency to fall in the rotarod test, balance time, and protein level of PSD-95 and dopamine receptor D1 in the 6-OHDA + marijuana group. These results suggest that marijuana may be helpful for motor disorders and synaptic changes in patients with Parkinson's disease.


Subject(s)
Behavior, Animal/drug effects , Cannabinoid Receptor Agonists/pharmacology , Disks Large Homolog 4 Protein/drug effects , Dronabinol/pharmacology , Dyskinesia, Drug-Induced/drug therapy , Medical Marijuana/pharmacology , Neuronal Plasticity/drug effects , Receptors, Dopamine D1/drug effects , Adrenergic Agents/pharmacology , Animals , Cannabinoid Receptor Agonists/administration & dosage , Disease Models, Animal , Dronabinol/administration & dosage , Male , Medical Marijuana/administration & dosage , Oxidopamine/pharmacology , Parkinson Disease/drug therapy , Plant Extracts , Rats , Rats, Wistar
5.
Article in English | MEDLINE | ID: mdl-32828853

ABSTRACT

A converging line of evidence is indicating that cannabinoids may have an opioid-sparing effect. This property, well validated in preclinical studies, allow when both drugs are co-administered to reduce the dose of opioids without loss of analgesic effects. A meta-analysis of pre-clinical studies indicated in 2017 that the median effective dose (ED50) of morphine administered in combination with delta-9-tetrahydrocannabinol (delta-9-THC) is 3.6 times lower than the ED50 of morphine alone (Nielsen et al., 2017). However, very few studies have been conducted in humans to validate this effect. This narrative review provides an update on whether or not cannabinoid drugs can be used to produce an opioid sparing effect. For this, various lines of evidence ranging from preclinical, epidemiological and human studies will be summarized. Overall, this review indicates that the preclinical results are strongly and consistently supportive of the presence of an opioid sparing effect of cannabinoid drugs. However, to date the clinical studies have been mostly negative; and, the evidence collected in humans so far is so limited that it is premature to conclude. Therefore, prospective high quality controlled clinical trials are still required to validate this. Priorities for future research are also discussed.


Subject(s)
Analgesics, Opioid/administration & dosage , Cannabinoids/administration & dosage , Clinical Trials as Topic/methods , Pain/drug therapy , Animals , Clinical Trials as Topic/standards , Dronabinol/administration & dosage , Drug Evaluation, Preclinical/methods , Drug Synergism , Humans , Morphine/administration & dosage , Opioid-Related Disorders/diagnosis , Opioid-Related Disorders/epidemiology , Opioid-Related Disorders/prevention & control , Pain/diagnosis , Pain/epidemiology
6.
Pharmazie ; 75(10): 463-469, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33305718

ABSTRACT

Cannabidiol (CBD) is an alkaloid present in Cannabis sativa, along with tetrahydrocannabinol (THC) and more than 100 other substances belonging to a group of compounds called cannabinoids. Whereas the legal status and medical use of Cannabis is a controversial issue in many countries, inconsistent legislation makes CBD status even more complicated. Some CBD products are legal in some countries, while banned in other countries, further compounding the confusion. In 2018, the Food and Drug Administration (FDA) approved the first CBD containing medical product, Epidiolex®, for the treatment of paediatric seizures. Currently, several clinical trials are in progress for the potential treatment of neurologic and behavioural disorders. CBD's current legal and regulatory status is a continuously evolving issue; the current review is presenting historical and present information regarding the use of CBD products worldwide.


Subject(s)
Cannabidiol/administration & dosage , Cannabis/chemistry , Medical Marijuana/administration & dosage , Animals , Cannabinoids/administration & dosage , Dronabinol/administration & dosage , Drug Approval/legislation & jurisprudence , Humans , Legislation, Drug , Marijuana Use/legislation & jurisprudence
7.
Best Pract Res Clin Anaesthesiol ; 34(3): 463-477, 2020 Sep.
Article in English | MEDLINE | ID: mdl-33004159

ABSTRACT

Chronic pain can be recurrent or constant pain that lasts for longer than 3 months and can result in disability, suffering, and a physical disturbance. Related to the complex nature of chronic pain, treatments have a pharmacological and non-pharmacological approach. Due to the opioid epidemic, alternative therapies have been introduced, and components of the plant Cannabis Sativa, Δ9-tetrahydrocannabinol (THC) and cannabidiol (CBD) have gained recent interest as a choice of treatment. The exact mechanism for CBD is currently unknown, but unlike the CBD's psychoactive counterpart, THC, the side effects of CBD itself have been shown to be overall much more benign. The current pharmaceutical products for the treatment of chronic pain are known as nabiximols, and they contain a ratio of THC combined with CBD, which has been promising. This review focuses on the treatment efficacy of CBD, THC: CBD-based treatments for chronic pain and adverse events with each.


Subject(s)
Analgesics/administration & dosage , Cannabidiol/administration & dosage , Cannabinoid Receptor Agonists/administration & dosage , Chronic Pain/drug therapy , Dronabinol/administration & dosage , Chronic Pain/diagnosis , Chronic Pain/physiopathology , Cross-Over Studies , Drug Administration Routes , Drug Combinations , Drug Therapy, Combination , Humans , Treatment Outcome
8.
J Pharmacol Toxicol Methods ; 106: 106937, 2020.
Article in English | MEDLINE | ID: mdl-33096236

ABSTRACT

INTRODUCTION: The assessment of the abuse potential of CNS-active drugs is a regulatory requirement. Drug discrimination is one of the nonclinical tests that contribute to this assessment by providing information on a drug's potential to induce a discriminative stimulus comparable to that of a known drug of abuse. AIM: The objective was to validate drug discrimination in the rat for the purpose of supporting regulatory submissions for novel drugs with potential cannabinoid-like activity. METHODS: Ten female Lister hooded rats were trained to discriminate no-drug from Δ9-THC (1.5 mg/kg, IP) under a FR10 schedule of reinforcement. Once trained, a Δ9-THC dose-response curve was obtained using doses of 0.25, 0.75, 1.5, and 3 mg/kg, IP. This was followed by evaluation of amphetamine (0.3 mg/kg, SC); morphine (3 mg/kg, IP); midazolam (2.5 mg/kg, PO); and the synthetic cannabinoids WIN55,212-2 (0.75 to 2 mg/kg, IP), CP-47,497 (0.5 to 2 mg/kg, IP), and JWH-018 (1 mg/kg, IP) for their discriminative stimulus similarity to Δ9-THC. RESULTS: Pharmacological specificity was demonstrated by achieving the anticipated dose-response curve for Δ9-THC, and a vehicle-like response for the non-cannabinoid drugs. Although full generalisation was obtained for JWH-018, in contrast to published literature, WIN55,212-2 and CP-47,497 failed to generalise to Δ9-THC. DISCUSSION: Based on the literature review performed in light of the results obtained, contrasting and unpredictable behavioural responses produced by cannabinoids in animals and humans raises the question of the reliability and relevance of including drug discrimination and self-administration studies within an abuse potential assessment for novel cannabinoid-like drugs.


Subject(s)
Discrimination, Psychological/drug effects , Dronabinol/adverse effects , Substance-Related Disorders/prevention & control , Amphetamine/administration & dosage , Amphetamine/adverse effects , Animals , Benzoxazines/administration & dosage , Benzoxazines/adverse effects , Cyclohexanols/administration & dosage , Cyclohexanols/adverse effects , Disease Models, Animal , Dose-Response Relationship, Drug , Dronabinol/administration & dosage , Drug Evaluation, Preclinical/methods , Female , Humans , Indoles/administration & dosage , Indoles/adverse effects , Injections, Intraperitoneal , Midazolam/administration & dosage , Midazolam/adverse effects , Morphine/administration & dosage , Morphine/adverse effects , Morpholines/administration & dosage , Morpholines/adverse effects , Naphthalenes/administration & dosage , Naphthalenes/adverse effects , Rats , Reinforcement, Psychology , Reproducibility of Results , Self Medication , Substance-Related Disorders/diagnosis , Substance-Related Disorders/etiology
9.
JAMA Psychiatry ; 77(8): 787-796, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32520316

ABSTRACT

Importance: The rapidly growing legal cannabis market includes new and highly potent products, the effects of which, to our knowledge, have not previously been examined in biobehavioral research studies because of federal restrictions on cannabis research. Objective: To use federally compatible, observational methods to study high-∆9-tetrahydrocannabinol (THC) legal market forms of cannabis. Design, Setting, and Participants: In this cohort study with a between-groups design that was conducted in a community and university setting, cannabis flower users and concentrate users were randomly assigned to higher- vs lower-THC products within user groups. Participants completed a baseline and an experimental mobile laboratory assessment that included 3 points: before, immediately after, and 1 hour after ad libitum legal market flower and concentrate use. Of the 133 individuals enrolled and assessed, 55 regular flower cannabis users (41.4%) and 66 regular concentrate cannabis users (49.6%) complied with the study's cannabis use instructions and had complete data across primary outcomes. Exposures: Flower users were randomly assigned to use either 16% or 24% THC flower and concentrate users were randomly assigned to use either 70% or 90% THC concentrate that they purchased from a dispensary. Main Outcomes and Measures: Primary outcome measures included plasma cannabinoids, subjective drug intoxication, and neurobehavioral tasks testing attention, memory, inhibitory control, and balance. Results: A total of 121 participants completed the study for analysis: 55 flower users (mean [SD] age, 28.8 [8.1] years; 25 women [46%]) and 66 concentrate users (mean [SD] age, 28.3 [10.4] years; 30 women [45%]). Concentrate users compared with flower users exhibited higher plasma THC levels and 11-hydroxyΔ9-THC (THC's active metabolite) across all points. After ad libitum cannabis administration, mean plasma THC levels were 0.32 (SE = 0.43) µg/mL in concentrate users (to convert to millimoles per liter, multiply by 3.18) and 0.14 (SE = 0.16) µg/mL in flower users. Most neurobehavioral measures were not altered by short-term cannabis consumption. However, delayed verbal memory (F1,203 = 32.31; P < .001) and balance function (F1,203 = 18.88; P < .001) were impaired after use. Differing outcomes for the type of product (flower vs concentrate) or potency within products were not observed. Conclusions and Relevance: This study provides information about the association of pharmacological and neurobehavioral outcomes with legal market cannabis. Short-term use of concentrates was associated with higher levels of THC exposure. Across forms of cannabis and potencies, users' domains of verbal memory and proprioception-focused postural stability were primarily associated with THC administration.


Subject(s)
Cannabis/adverse effects , Cognitive Dysfunction/chemically induced , Dronabinol/analogs & derivatives , Dronabinol/adverse effects , Dronabinol/blood , Flowers/adverse effects , Plant Extracts/adverse effects , Sensation Disorders/chemically induced , Adult , Attention/drug effects , Dronabinol/administration & dosage , Executive Function/drug effects , Female , Humans , Inhibition, Psychological , Male , Plant Extracts/administration & dosage , Postural Balance/drug effects , Verbal Learning/drug effects , Young Adult
10.
Drug Metab Dispos ; 48(10): 1008-1017, 2020 10.
Article in English | MEDLINE | ID: mdl-32587099

ABSTRACT

Cannabis is used for both recreational and medicinal purposes. The most abundant constituents are the cannabinoids - cannabidiol (CBD, nonpsychoactive) and (-)-trans-Δ9-tetrahydrocannabinol (THC, psychoactive). Both have been reported to reversibly inhibit or inactivate cytochrome P450 (CYPs) enzymes. However, the low aqueous solubility, microsomal protein binding, and nonspecific binding to labware were not considered, potentially leading to an underestimation of CYPs inhibition potency. Therefore, the binding-corrected reversible (IC50,u) and irreversible (K I,u ) inhibition potency of each cannabinoid toward major CYPs were determined. The fraction unbound of CBD and THC in the incubation mixture was 0.12 ± 0.04 and 0.05 ± 0.02, respectively. The IC50,u for CBD toward CYP1A2, 2C9, 2C19, 2D6, and 3A was 0.45 ± 0.17, 0.17 ± 0.03, 0.30 ± 0.06, 0.95 ± 0.50, and 0.38 ± 0.11 µM, respectively; the IC50,u for THC was 0.06 ± 0.02, 0.012 ± 0.001, 0.57 ± 0.22, 1.28 ± 0.25, and 1.30 ± 0.34 µM, respectively. Only CBD showed time-dependent inactivation (TDI) of CYP1A2, 2C19, and CYP3A, with inactivation efficiencies (k inact/K I,u) of 0.70 ± 0.34, 0.11 ± 0.06, and 0.14 ± 0.04 minutes-1 µM-1, respectively. A combined (reversible inhibition and TDI) mechanistic static model populated with these data predicted a moderate to strong pharmacokinetic interaction risk between orally administered CBD and drugs extensively metabolized by CYP1A2/2C9/2C19/2D6/3A and between orally administered THC and drugs extensively metabolized by CYP1A2/2C9/3A. These predictions will be extended to a dynamic model using physiologically based pharmacokinetic modeling and simulation and verified with a well-designed clinical cannabinoid-drug interaction study. SIGNIFICANCE STATEMENT: This study is the first to consider the impact of limited aqueous solubility, nonspecific binding to labware, or extensive binding to incubation protein shown by cannabidiol (CBD) and delta-9-tetrahydrocannabinol (THC) on their true cytochrome P450 inhibitory potency. A combined mechanistic static model predicted a moderate to strong pharmacokinetic interaction risk between orally administered CBD and drugs extensively metabolized by CYP1A2, 2C9, 2C19, 2D6, or 3A and between orally administered THC and drugs extensively metabolized by CYP1A2, 2C9, or 3A.


Subject(s)
Cannabidiol/pharmacokinetics , Cytochrome P-450 Enzyme Inhibitors/pharmacokinetics , Dronabinol/pharmacokinetics , Administration, Oral , Adult , Cannabidiol/administration & dosage , Cytochrome P-450 Enzyme Inhibitors/administration & dosage , Cytochrome P-450 Enzyme System/metabolism , Dronabinol/administration & dosage , Drug Evaluation, Preclinical , Drug Interactions , Humans , Inhibitory Concentration 50 , Microsomes, Liver
11.
Medicina (Kaunas) ; 56(5)2020 May 15.
Article in English | MEDLINE | ID: mdl-32429074

ABSTRACT

Cannabis has been used for centuries for therapeutic purposes. In the last century, the plant was demonized due to its high abuse liability and supposedly insufficient health benefits. However, recent decriminalization policies and new scientific evidence have increased the interest in cannabis therapeutic potential of cannabis and paved the way for the release of marketing authorizations for cannabis-based products. Although several synthetic and standardized products are currently available on the market, patients' preferences lean towards herbal preparations, because they are easy to handle and self-administer. A literature search was conducted on multidisciplinary research databases and international agencies or institutional websites. Despite the growing popularity of medical cannabis, little data is available on the chemical composition and preparation methods of medical cannabis extracts. The authors hereby report the most common cannabis preparations, presenting their medical indications, routes of administration and recommended dosages. A practical and helpful guide for prescribing doctors is provided, including suggested posology, titration strategies and cannabinoid amounts in herbal preparations obtained from different sources of medical cannabis.


Subject(s)
Cannabidiol/administration & dosage , Dronabinol/administration & dosage , Medical Marijuana/administration & dosage , Cannabidiol/adverse effects , Cannabidiol/pharmacology , Dronabinol/adverse effects , Dronabinol/pharmacology , Herbal Medicine , Humans , Medical Marijuana/adverse effects , Medical Marijuana/pharmacology , Plant Preparations/therapeutic use
12.
Eur Neuropsychopharmacol ; 36: 206-216, 2020 07.
Article in English | MEDLINE | ID: mdl-32273144

ABSTRACT

The endogenous opioid and cannabinoid receptor systems are widely distributed and co-localized throughout central and peripheral nervous system regions. A large body of preclinical evidence suggests that there are functional interactions between these two systems that may be leveraged to address various health conditions. Numerous animal studies have shown that cannabinoid agonists (e.g., delta-9-tetrahydrocannabinol [Δ9-THC]) enhance the analgesic effects of µ-opioid analgesics as evidenced by decreasing the opioid dose required for analgesia (i.e., opioid sparing) and extending the duration of the opioid analgesia. In contrast, controlled human laboratory studies and clinical trials have not demonstrated robust analgesic or opioid-sparing effects from opioid-cannabinoid combinations. Meta-analyses of the literature (clinical trials, controlled laboratory studies; some non-controlled studies/case reports) have examined the effects of cannabis/cannabinoids for pain relief in those taking a wide variety of analgesics, including prescription opioid medications. These data do not strongly support the use of cannabinoids for chronic pain nor do prospective studies demonstrate significant cannabinoid-mediated opioid-sparing effects. Preclinical studies have also suggested a role for cannabinoids for the treatment of opioid withdrawal. Controlled laboratory and clinical studies suggest that there may be a modest signal for Δ9-THC to suppress some opioid signs and symptoms but they are not completely ameliorated and there may also be concerns around safety of Δ9-THC administration in a state of heightened autonomic arousal as occurs with opioid withdrawal. Despite anecdotal and correlational reports suggesting a benefit of cannabis on reducing opioid overdose, there is no strong data supporting this contention and emerging reports have conflicting results. In summary, there is a groundswell of public advocacy supporting the use of cannabis and cannabinoids to replace opioid analgesics or to reduce opioid use; however, the extant controlled clinical data do not support the role of cannabinoids for opioid replacement or opioid-sparing effects when treating opioid use disorder or chronic pain.


Subject(s)
Analgesics, Opioid/administration & dosage , Cannabinoid Receptor Agonists/administration & dosage , Chronic Pain/drug therapy , Dronabinol/administration & dosage , Pain Management/methods , Animals , Chronic Pain/diagnosis , Chronic Pain/epidemiology , Clinical Trials as Topic/methods , Humans , Pain Management/trends , Prospective Studies
13.
Biomolecules ; 10(2)2020 02 11.
Article in English | MEDLINE | ID: mdl-32054131

ABSTRACT

The phytocannabinoid-based medicine Sativex® is currently marketed for the treatment of spasticity and pain in multiple sclerosis patients and is being investigated for other central and peripheral pathological conditions. It may also serve in Veterinary Medicine for the treatment of domestic animals, in particular for dogs affected by different pathologies, including human-like pathological conditions. With the purpose of assessing different dosing paradigms for using Sativex in Veterinary Medicine, we investigated its pharmacokinetics when administered to naïve dogs via sublingual delivery. In the single dose arm of the study, adult Beagle dogs were treated with 3 consecutive sprays of Sativex, and blood samples were collected at 12 intervals up to 24 h later. In the multiple dose arm of the study, Beagle dogs received 3 sprays daily for 14 days, and blood samples were collected for 24 h post final dose. Blood was used to obtain plasma samples and to determine the levels of cannabidiol (CBD), Δ9-tetrahydrocannabinol (Δ9-THC) and its metabolite 11-hydroxy-Δ9-THC. Maximal plasma concentrations of both Δ9-THC (Cmax = 18.5 ng/mL) and CBD (Cmax = 10.5 ng/mL) were achieved 2 h after administration in the single dose condition and at 1 h in the multiple dose treatment (Δ9-THC: Cmax = 24.5 ng/mL; CBD: Cmax = 15.2 ng/mL). 11hydroxy-Δ9-THC, which is mainly formed in the liver from Δ9-THC, was almost undetected, which is consistent with the use of sublingual delivery. A potential progressive accumulation of both CBD and Δ9-THC was detected following repeated exposure, with maximum plasma concentrations for both cannabinoids being achieved following multiple dose. Neurological status, body temperature, respiratory rate and some hemodynamic parameters were also recorded in both conditions, but in general, no changes were observed. In conclusion, this study demonstrates that single or multiple dose sublingual administration of Sativex to naïve dogs results in the expected pharmacokinetic profile, with maximal levels of phytocannabinoids detected at 1-2 h and suggested progressive accumulation after the multiple dose treatment.


Subject(s)
Analgesics/administration & dosage , Analgesics/pharmacokinetics , Cannabidiol/administration & dosage , Cannabidiol/pharmacokinetics , Dronabinol/administration & dosage , Dronabinol/pharmacokinetics , Administration, Sublingual , Analgesics/blood , Animals , Blood Pressure/drug effects , Body Temperature/drug effects , Cannabidiol/blood , Dog Diseases , Dogs , Dronabinol/blood , Drug Combinations , Female , Heart Rate/drug effects , Male , Oral Sprays , Plant Extracts/administration & dosage , Plant Extracts/blood , Plant Extracts/pharmacokinetics , Respiratory Rate/drug effects
14.
Am J Hosp Palliat Care ; 37(8): 589-593, 2020 Aug.
Article in English | MEDLINE | ID: mdl-31986898

ABSTRACT

BACKGROUND: Palliative medicine physicians are challenged by lack of guidance regarding effectiveness and dosing of cannabis products in the setting of their emerging popularity. OBJECTIVE: The aim of this study was to describe early patterns of tetrahydrocannabinol (THC) and cannabidiol (CBD) use in Florida following passage of the state's first medical marijuana law. We describe here the perceived benefits, side effects, and beliefs expressed by patients in a single outpatient academic palliative medicine practice. METHODS: A cross-sectional survey was performed of a sequential convenience sample of patients who presented to an outpatient academic palliative medicine clinic over a 3-month period. RESULTS: In all, 24% (14/58) of respondents reported THC use, with half using THC on a daily basis. Patients reported improvements in pain, appetite, and nausea. In all, 71% (10/14) began using THC after the diagnosis of their chronic illness, and the most common form of usage was vaping. In all, 24% (14/58) of patients reported CBD use. Patients reported improvements in pain, and the most common form of usage was topical application. None of the patients had used CBD prior to the onset of their chronic illness. In all, 21% (3/14) of THC users and 21% (3/14) of CBD users thought that their substance was helping to cure their illness. Individual reported side effects in both groups were minimal. CONCLUSIONS: Approximately a quarter of outpatient palliative care patients use THC or CBD, often on a daily basis. Palliative care providers should be aware of the frequency, diverse usage, and beliefs behind cannabis product use in this patient population.


Subject(s)
Cannabidiol/therapeutic use , Dronabinol/therapeutic use , Medical Marijuana/therapeutic use , Pain/drug therapy , Palliative Care/methods , Adult , Aged , Aged, 80 and over , Cannabidiol/administration & dosage , Cannabidiol/adverse effects , Cross-Sectional Studies , Dronabinol/administration & dosage , Dronabinol/adverse effects , Drug Administration Routes , Florida , Humans , Medical Marijuana/administration & dosage , Medical Marijuana/adverse effects , Middle Aged , Practice Patterns, Physicians'
15.
J Neurol ; 267(2): 415-421, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31655890

ABSTRACT

BACKGROUND: The symptomatic treatment of myotonia and myalgia in patients with dystrophic and non-dystrophic myotonias is often not satisfactory. Some patients anecdotally report symptoms' relief through consumption of cannabis. METHODS: A combination of cannabidiol and tetrahydrocannabinol (CBD/THC) was prescribed as compassionate use to six patients (four patients with myotonic dystrophy types 1 and 2, and 2 patients with CLCN1-myotonia) with therapy-resistant myotonia and myalgia. CBD/THC oil was administered on a low dose in the first 2 weeks and adjusted to a higher dose in the following 2 weeks. Myotonia behaviour scale (MBS), hand-opening time, visual analogue scales (VAS) for myalgia and myotonia, and fatigue and daytime sleepiness severity scale (FSS, ESS) were performed weekly to monitor treatment response. RESULTS: All patients reported an improvement of myotonia especially in weeks 3 and 4 of treatment: MBS improved of at least 2 points in all patients, the hand-opening time variously improved in 5 out of 6 patients. Chronic myalgia was reported by both DM2 patients at baseline, one of them experienced a significant improvement of myalgia under treatment. Some gastrointestinal complaints, as abdominal pain and diarrhoea, improved in 3 patients; however, 4 out of 6 patients reported new-onset constipation. No other relevant side effect was noticed. CONCLUSIONS: These first empirical results suggest a potentially beneficial role of CBD/THC in alleviating myotonia and should encourage further research in this field including a randomized-controlled trial on larger cohorts.


Subject(s)
Cannabidiol/pharmacology , Cannabinoid Receptor Modulators/pharmacology , Dronabinol/pharmacology , Muscular Dystrophies/drug therapy , Myalgia/drug therapy , Myotonia/drug therapy , Adult , Aged , Cannabidiol/administration & dosage , Cannabinoid Receptor Modulators/administration & dosage , Chronic Disease , Cohort Studies , Compassionate Use Trials , Dronabinol/administration & dosage , Drug Combinations , Female , Humans , Male , Middle Aged , Oils , Treatment Outcome
16.
Lancet ; 394(10214): 2073-2083, 2019 12 07.
Article in English | MEDLINE | ID: mdl-31711629

ABSTRACT

BACKGROUND: An ongoing outbreak of lung injury associated with e-cigarettes or vaping (also known as E-VALI or VALI) started in March, 2019, in the USA. The cause, diagnosis, treatment, and course of this disease remains unknown. METHODS: In this multicentre, prospective, observational, cohort study, we collected data on all patients with lung injury associated with e-cigarettes or vaping seen in Intermountain Healthcare, an integrated health system based in Utah, USA, between June 27 and Oct 4, 2019. Telecritical care, based in Salt Lake City, UT, USA, was used as the central repository for case validation, public reporting, and system-wide dissemination of expertise, which included a proposed diagnosis and treatment guideline for lung injury associated with e-cigarettes or vaping. We extracted data on patient presentation, treatment, and short-term follow-up (2 weeks after discharge) from chart review and interviews with patients undertaken by the Utah Department of Health (Salt Lake City, UT, USA). FINDINGS: 60 patients presented with lung injury associated with e-cigarettes or vaping at 13 hospitals or outpatient clinics in the integrated health system. 33 (55%) of 60 were admitted to an intensive care unit (ICU). 53 (88%) of 60 patients presented with constitutional symptoms, 59 (98%) with respiratory symptoms, and 54 (90%) with gastrointestinal symptoms. 54 (90%) of 60 were given antibiotics and 57 (95%) were given steroids. Six (10%) of 60 patients were readmitted to an ICU or hospital within 2 weeks, three (50%) of whom had relapsed with vaping or e-cigarette use. Of 26 patients who were followed up within 2 weeks, despite clinical and radiographic improvement in all, many had residual abnormalities on chest radiographs (ten [67%] of 15) and pulmonary function tests (six [67%] of nine). Two patients died and lung injury associated with e-cigarettes or vaping was thought to be a contributing factor, but not the cause of death, for both. INTERPRETATION: Lung injury associated with e-cigarettes or vaping is an emerging illness associated with severe lung injury and constitutional and gastrointestinal symptoms. Increased awareness has led to identification of a broad spectrum of severity of illness in patients who were treated with antibiotics and steroids. Despite improvement, at short-term follow-up many patients had residual abnormalities. Lung injury associated with e-cigarettes or vaping remains a clinical diagnosis with symptoms that overlap infectious and other lung diseases. Maintaining a high index of suspicion for this disease is important as work continues in understanding the cause or causes, optimal therapy, and long-term outcomes of these patients. FUNDING: Intermountain Healthcare.


Subject(s)
Acute Lung Injury/etiology , Vaping/adverse effects , Acute Lung Injury/diagnostic imaging , Acute Lung Injury/epidemiology , Acute Lung Injury/therapy , Adult , Anti-Bacterial Agents/therapeutic use , Bronchoscopy , Cannabidiol/administration & dosage , Cohort Studies , Disease Outbreaks , Dronabinol/administration & dosage , Electronic Nicotine Delivery Systems , Female , Glucocorticoids/therapeutic use , Humans , Length of Stay , Male , Nicotine/administration & dosage , Noninvasive Ventilation , Oxygen Inhalation Therapy , Prospective Studies , Respiration, Artificial , Tomography, X-Ray Computed , Utah/epidemiology , Young Adult
17.
Eur J Gastroenterol Hepatol ; 31(11): 1376-1381, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31567639

ABSTRACT

OBJECTIVE: Use of medical cannabis for improving symptoms of inflammatory bowel disease is increasing. However, reports on long-term outcomes are lacking. This prospective, observational study assessed the effects of licensed cannabis use among patients with inflammatory bowel disease. METHODS: Dose and mode of consumption, adverse events, use of other medications, and long-term effects were evaluated among 127 patients with inflammatory bowel disease using legalized medical cannabis. Blood count, albumin, and C-reactive protein were assessed before, 1 month, and at least 1 year after medical cannabis therapy was initiated. Questionnaires on disease activity, patient function, and signs of addiction were completed by patients and by a significant family member to assess its effects. RESULTS: The average dose used was 31 ± 15 g/month. The average Harvey-Bradshaw index improved from 14 ± 6.7 to 7 ± 4.7 (P < 0.001) during a median follow-up of 44 months (interquartile range, 24-56 months). There was a slight, but statistically significant, average weight gain of 2 kg within 1 year of cannabis use. The need for other medications was significantly reduced. Employment among patients increased from 65 to 74% (P < 0.05). We conclude that the majority of inflammatory bowel disease patients using cannabis are satisfied with a dose of 30 g/month. We did not observe negative effects of cannabis use on the patients' social or occupational status. CONCLUSIONS: Cannabis use by inflammatory bowel disease patients can induce clinical improvement and is associated with reduced use of medication and slight weight gain. Most patients respond well to a dose of 30 g/month, or 21 mg Δ9-tetra- hydrocannabinol (THC) and 170 mg Cannabidiol (CBD) per day.


Subject(s)
Cannabidiol/administration & dosage , Dronabinol/administration & dosage , Inflammatory Bowel Diseases/drug therapy , Medical Marijuana/therapeutic use , Adolescent , Adult , Aged , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Biological Products/therapeutic use , C-Reactive Protein/metabolism , Drug Administration Routes , Employment , Female , Humans , Immunologic Factors/therapeutic use , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/physiopathology , Israel , Male , Mesalamine/therapeutic use , Middle Aged , Patient Reported Outcome Measures , Patient Satisfaction , Prospective Studies , Serum Albumin/metabolism , Treatment Outcome , Tumor Necrosis Factor Inhibitors/therapeutic use , Weight Gain , Young Adult
18.
Seizure ; 72: 23-27, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31550641

ABSTRACT

PURPOSE: Interest in the use of artisanal cannabinoids in pediatric epilepsy has increased but safety and utility data are lacking. Our aim was to prospectively characterize the use of oral cannabis extracts (OCE) in a refractory pediatric epilepsy population. METHODS: Families considering the use of an OCE were enrolled in a prospective observational study. Baseline seizure frequency was assessed over a period of 4 weeks. Seizure frequency, CBD and THC-COOH levels were assessed every 4 weeks during a 12-week treatment period. Response was defined as at least a 50% reduction in seizure frequency over the final 8 weeks of the study relative to baseline. RESULTS: Consent was obtained in 32 children; 11 were excluded from analysis (3 failed to complete baseline data, 3 started OCE before completing baseline period and 5 did not start OCE) leaving 21 to be included in subsequent analyses. Median age was 10.3 years (IQR 6.8-12.6), 13 (62%) were male and median seizure frequency was 2.7 seizures/day during the baseline period. The median of the high dose of CBD that was administered during the observation period was of 0.9 (0.6-2.2) mg/kg/day. Of the 21 subjects who were included in the analysis, 5 (24%) were responders. OCE was stopped early in 3 subjects (14%) due to a perceived increase in seizures. THC-COOH and CBD blood levels did not have a significant association with response status (p = 0.95 CBD, p = 0.53 THC-COOH, N = 14). CONCLUSION: The observed response rate in this study is similar to placebo rates in prospective randomized trials of pharmaceutical grade products and the withdrawal rate is greater than rates obtained with retrospective methods. Doses of OCE administered were lower than doses used in randomized trials.


Subject(s)
Anticonvulsants/administration & dosage , Cannabis , Drug Resistant Epilepsy/diagnosis , Drug Resistant Epilepsy/drug therapy , Medical Marijuana/administration & dosage , Plant Extracts/administration & dosage , Administration, Oral , Adolescent , Cannabidiol/administration & dosage , Child , Child, Preschool , Dronabinol/administration & dosage , Female , Humans , Male , Prospective Studies
19.
Pharmacol Biochem Behav ; 184: 172741, 2019 09.
Article in English | MEDLINE | ID: mdl-31336109

ABSTRACT

RATIONALE: Cannabidiol (CBD), a compound found in many strains of the Cannabis genus, is increasingly available in e-cigarette liquids as well as other products. CBD use has been promoted for numerous purported benefits which have not been rigorously assessed in preclinical studies. OBJECTIVE: To further validate an inhalation model to assess CBD effects in the rat. The primary goal was to determine plasma CBD levels after vapor inhalation and compare that with the levels observed after injection. Secondary goals were to determine if hypothermia is produced in male Sprague-Dawley rats and if CBD affects nociception measured by the warm water tail-withdrawal assay. METHODS: Blood samples were collected from rats exposed for 30 min to vapor generated by an e-cigarette device using CBD (100, 400 mg/mL in the propylene glycol vehicle). Separate experiments assessed the body temperature response to CBD in combination with nicotine (30 mg/mL) and the anti-nociceptive response to CBD. RESULTS: Vapor inhalation of CBD produced concentration-related plasma CBD levels in male and female Wistar rats that were within the range of levels produced by 10 or 30 mg/kg, CBD, i.p. Dose-related hypothermia was produced by CBD in male Sprague-Dawley rats, and nicotine (30 mg/mL) inhalation enhanced this effect. CBD inhalation had no effect on anti-nociception alone or in combination with Δ9-tetrahydrocannabinol inhalation. CONCLUSIONS: The vapor-inhalation approach is a suitable pre-clinical model for the investigation of the effects of inhaled CBD. This route of administration produces hypothermia in rats, while i.p. injection does not, at comparable plasma CBD levels.


Subject(s)
Cannabidiol/administration & dosage , Cannabidiol/pharmacology , E-Cigarette Vapor/pharmacology , Plant Extracts/administration & dosage , Plant Extracts/pharmacology , Administration, Inhalation , Animals , Body Temperature/drug effects , Cannabidiol/blood , Cannabis/chemistry , Cohort Studies , Dose-Response Relationship, Drug , Dronabinol/administration & dosage , Dronabinol/pharmacology , Electronic Nicotine Delivery Systems , Female , Hypothermia/chemically induced , Male , Models, Animal , Nicotine/administration & dosage , Nicotine/pharmacology , Nociception/drug effects , Plant Extracts/blood , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptor, Serotonin, 5-HT1A/metabolism
20.
Pharmacol Biochem Behav ; 184: 172739, 2019 09.
Article in English | MEDLINE | ID: mdl-31283908

ABSTRACT

The high prevalence of concomitant cannabis and nicotine use has implications for sensory and cognitive processing. While nicotine tends to enhance function in these domains, cannabis use has been associated with both sensory and cognitive impairments, though the underlying mechanisms are unclear. Additionally, the interaction of the nicotinic (nAChR) and cannabinoid (CB1) receptor systems has received limited study in terms of sensory/cognitive processes. This study involving healthy volunteers assessed the acute separate and combined effects of nabilone (a CB1 agonist) and nicotine on sensory processing as assessed by auditory deviance detection and indexed by the mismatch negativity (MMN) event-related potential. It was hypothesized that nabilone would impair auditory discriminability as shown by diminished MMN amplitudes, but not when administered in combination with nicotine. 20 male non-smokers and non-cannabis-users were assessed using a 5-stimulus 'optimal' multi-feature MMN paradigm within a randomized, placebo controlled design (placebo; nabilone [0.5 mg]; nicotine [6 mg]; and nicotine + nabilone). Treatment effects were region- and deviant-dependent. At the temporal regions (mastoid sites), MMN was reduced by nabilone and nicotine separately, whereas co-administration resulted in no impairment. At the frontal region, MMN was enhanced by co-administration of nicotine and nabilone, with no MMN effects being found with separate treatment. These neural effects have relevance for sensory/cognitive processes influenced by separate and simultaneous use of cannabis and tobacco and may have treatment implications for disorders associated with sensory dysfunction and impairments in endocannabinoid and nicotinic cholinergic neurotransmission.


Subject(s)
Cannabinoid Receptor Agonists/pharmacology , Dronabinol/analogs & derivatives , Evoked Potentials, Auditory/drug effects , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Acoustic Stimulation/methods , Adult , Cannabinoid Receptor Agonists/administration & dosage , Double-Blind Method , Dronabinol/administration & dosage , Dronabinol/pharmacology , Drug Therapy, Combination/methods , Electroencephalography/methods , Electrooculography/methods , Frontal Lobe/drug effects , Healthy Volunteers , Humans , Male , Nicotine/administration & dosage , Nicotinic Agonists/administration & dosage , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/metabolism , Receptors, Nicotinic/metabolism , Schizophrenia/drug therapy , Temporal Lobe/drug effects , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL