Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 930
Filter
2.
CNS Drugs ; 38(8): 597-612, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38951463

ABSTRACT

Synthetic cannabinoids are compounds made in the laboratory to structurally and functionally mimic phytocannabinoids from the Cannabis sativa L. plant, including delta-9-tetrahydrocannabinol (THC). Synthetic cannabinoids (SCs) can signal via the classical endogenous cannabinoid system (ECS) and the greater endocannabidiome network, highlighting their signalling complexity and far-reaching effects. Dronabinol and nabilone, which mimic THC signalling, have been approved by the Food and Drug Administration (FDA) for treating nausea associated with cancer chemotherapy and/or acquired immunodeficiency syndrome (AIDS). However, there is ongoing interest in these two drugs as potential analgesics for a variety of other clinical conditions, including neuropathic pain, spasticity-related pain, and nociplastic pain syndromes including fibromyalgia, osteoarthritis, and postoperative pain, among others. In this review, we highlight the signalling mechanisms of FDA-approved synthetic cannabinoids, discuss key clinical trials that investigate their analgesic potential, and illustrate challenges faced when bringing synthetic cannabinoids to the clinic.


Subject(s)
Cannabinoids , Pain , Humans , Cannabinoids/pharmacology , Cannabinoids/therapeutic use , Cannabinoids/adverse effects , Cannabinoids/chemical synthesis , Pain/drug therapy , Animals , Analgesics/pharmacology , Analgesics/therapeutic use , Dronabinol/pharmacology , Dronabinol/therapeutic use , Synthetic Drugs/pharmacology , Synthetic Drugs/therapeutic use
3.
Ugeskr Laeger ; 186(15)2024 Apr 08.
Article in Danish | MEDLINE | ID: mdl-38708699

ABSTRACT

In the last decade, patients with chronic pain have expressed increasing interest in cannabis-derived products for adjuvant therapy when treatment is deemed refractory to conventional analgesics. At present, clinical evidence to support this treatment approach appears to be sparse. Not because clinical studies as such are lacking, but rather as a result of methodological bias in relation to study design, patient populations, and treatment protocols. In this review, research in cannabis medicine for relief of chronic pain is reviewed, mainly with reference to published meta-analytic studies.


Subject(s)
Chronic Pain , Medical Marijuana , Humans , Chronic Pain/drug therapy , Medical Marijuana/therapeutic use , Medical Marijuana/adverse effects , Dronabinol/therapeutic use , Analgesics/therapeutic use
4.
Trials ; 25(1): 293, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38693590

ABSTRACT

BACKGROUND: Distressing symptoms are common in advanced cancer. Medicinal cannabinoids are commonly prescribed for a variety of symptoms. There is little evidence to support their use for most indications in palliative care. This study aims to assess a 1:20 delta-9-tetrahydrocannabinol/cannabidiol (THC/CBD) cannabinoid preparation in the management of symptom distress in patients with advanced cancer undergoing palliative care. METHODS AND DESIGN: One hundred and fifty participants will be recruited across multiple sites in Queensland, Australia. A teletrial model will facilitate the recruitment of patients outside of major metropolitan areas. The study is a pragmatic, multicenter, randomised, placebo-controlled, two-arm trial of escalating doses of an oral 1:20 THC/CBD medicinal cannabinoid preparation (10 mg THC:200 mg CBD/mL). It will compare the efficacy and safety outcomes of a titrated dose range of 2.5 mg THC/50mgCBD to 30 mg THC/600 mg CBD per day against a placebo. There is a 2-week patient-determined titration phase, to reach a dose that achieves symptom relief or intolerable side effects, with a further 2 weeks of assessment on the final dose. The primary objective is to assess the effect of escalating doses of a 1:20 THC/CBD medicinal cannabinoid preparation against placebo on change in total symptom distress score, with secondary objectives including establishing a patient-determined effective dose, the effect on sleep quality and overall quality of life. Some patients will be enrolled in a sub-study which will more rigorously evaluate the effect on sleep. DISCUSSION: MedCan-3 is a high-quality, adequately powered, placebo-controlled trial which will help demonstrate the utility of a THC:CBD 1:20 oral medicinal cannabis product in reducing total symptom distress in this population. Secondary outcomes may lead to new hypotheses regarding medicinal cannabis' role in particular symptoms or in particular cancers. The sleep sub-study will test the feasibility of using actigraphy and the Insomnia Severity Index (ISI) in this cohort. This will be the first large-scale palliative care randomised clinical trial to utilise the teletrial model in Australia. If successful, this will have significant implications for trial access for rural and remote patients in Australia and internationally. TRIAL REGISTRATION: ANZCTR ACTRN12622000083796 . Protocol number 001/20. Registered on 21 January 2022. Recruitment started on 8 August 2022.


Subject(s)
Cannabidiol , Dronabinol , Medical Marijuana , Neoplasms , Palliative Care , Humans , Administration, Oral , Cannabidiol/administration & dosage , Cannabidiol/adverse effects , Cannabidiol/therapeutic use , Double-Blind Method , Dronabinol/therapeutic use , Dronabinol/administration & dosage , Drug Combinations , Medical Marijuana/therapeutic use , Medical Marijuana/adverse effects , Medical Marijuana/administration & dosage , Multicenter Studies as Topic , Neoplasms/drug therapy , Neoplasms/complications , Palliative Care/methods , Quality of Life , Queensland , Randomized Controlled Trials as Topic , Symptom Burden , Time Factors , Treatment Outcome
5.
Epilepsia ; 65(8): 2270-2279, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38808601

ABSTRACT

OBJECTIVE: Cannabidiol-enriched oil (CBDO) is being used increasingly to improve seizure control in adult patients with drug-resistant epilepsy (DRE), despite the lack of large-scale studies supporting its efficacy in this patient population. We aimed to assess the effects of add-on CBDO on seizure frequency as well as on gait, cognitive, affective, and sleep-quality metrics, and to explore the electrophysiological changes in responder and non-responder DRE patients treated with add-on CBDO. METHODS: We prospectively recruited adult DRE patients who were treated with add-on CBDO. Patients were evaluated prior to treatment and following 4 weeks of a maintenance daily dose of ≈260 mg CBD and ≈12 mg Δ9-tetrahydrocannabinol (THC). The outcome measures included seizure response to CBDO (defined as ≥50% decrease in seizures compared to pre-CBDO baseline), gait testing, Montreal Cognitive Assessment (MoCA), Hospital Anxiety and Depression Scale (HADS), and sleep-quality questionnaire assessments. Patients underwent electroencephalography (EEG) recording during rest as well as event-related potentials (ERPs) during visual Go/NoGo task while sitting and while walking. RESULTS: Nineteen patients were recruited, of which 16 finished pre- and post-CBDO assessments. Seven patients (43.75%) were responders demonstrating an average reduction of 82.4% in seizures, and nine patients (56.25%) were non-responders with an average seizure increase of 30.1%. No differences in demographics and clinical parameters were found between responders and non-responders at baseline. However, responders demonstrated better performance in the dual-task walking post-treatment (p = .015), and correlation between increase in MoCA and seizure reduction (r = .810, p = .027). Post-CBDO P300 amplitude was lower during No/Go-sitting in non-responders (p = .028) and during No/Go-walking in responders (p = .068). SIGNIFICANCE: CBDO treatment can reduce seizures in a subset of patients with DRE, but could aggravate seizure control in a minority of patients; yet we found no specific baseline clinical or electrophysiological characteristics that are associated with response to CBDO. However, changes in ERPs in response to treatment could be a promising direction to better identify patients who could benefit from CBDO treatment.


Subject(s)
Anticonvulsants , Cannabidiol , Drug Resistant Epilepsy , Electroencephalography , Humans , Male , Cannabidiol/therapeutic use , Female , Drug Resistant Epilepsy/drug therapy , Drug Resistant Epilepsy/physiopathology , Adult , Prospective Studies , Anticonvulsants/therapeutic use , Middle Aged , Young Adult , Evoked Potentials/drug effects , Evoked Potentials/physiology , Treatment Outcome , Dronabinol/therapeutic use , Gait/drug effects , Gait/physiology , Oils
6.
Int J Mol Sci ; 25(8)2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38673788

ABSTRACT

Phytocannabinoids, a diverse group of naturally occurring compounds extracted from the Cannabis plant, have attracted interest due to their potential pharmacological effects and medicinal uses. This comprehensive review presents the intricate pharmacological profiles of phytocannabinoids while exploring the diverse impacts these substances have on biological systems. From the more than one hundred cannabinoids which were identified in the Cannabis plant so far, cannabidiol (CBD) and tetrahydrocannabinol (THC) are two of the most extensively studied phytocannabinoids. CBD is a non-psychoactive compound, which exhibits potential anti-inflammatory, neuroprotective, and anxiolytic properties, making it a promising candidate for a wide array of medical conditions. THC, known for its psychoactive effects, possesses analgesic and antiemetic properties, contributing to its therapeutic potential. In addition to THC and CBD, a wide range of additional phytocannabinoids have shown intriguing pharmacological effects, including cannabichromene (CBC), cannabigerol (CBG), and cannabinol (CBN). The endocannabinoid system, made up of the enzymes involved in the production and breakdown of endocannabinoids, cannabinoid receptors (CB1 and CB2), and endogenous ligands (endocannabinoids), is essential for preserving homeostasis in several physiological processes. Beyond their effects on the endocannabinoid system, phytocannabinoids are studied for their ability to modify ion channels, neurotransmitter receptors, and anti-oxidative pathways. The complex interaction between phytocannabinoids and biological systems offers hope for novel treatment approaches and lays the groundwork for further developments in the field of cannabinoid-based medicine. This review summarizes the state of the field, points out information gaps, and emphasizes the need for more studies to fully realize the therapeutic potential of phytocannabinoids.


Subject(s)
Cannabinoids , Humans , Cannabinoids/therapeutic use , Cannabinoids/pharmacology , Animals , Cannabis/chemistry , Endocannabinoids/metabolism , Endocannabinoids/therapeutic use , Cannabidiol/therapeutic use , Cannabidiol/pharmacology , Phytochemicals/therapeutic use , Phytochemicals/pharmacology , Dronabinol/therapeutic use , Dronabinol/pharmacology
7.
CNS Drugs ; 38(5): 375-385, 2024 May.
Article in English | MEDLINE | ID: mdl-38597988

ABSTRACT

BACKGROUND: Disordered autonomic nervous system regulation and supraspinal pain inhibition have been repeatedly described in chronic pain. We aimed to explore the effects of δ-9-tetrahydrocannabinol (THC), an emerging treatment option, on autonomic nervous system and central pain modulation measures in patients with chronic pain. METHODS: Twelve male patients with chronic radicular neuropathic pain participated in a randomized, double-blind, crossover, placebo-controlled, single-administration trial. Low/high frequency (LF/HF) heart rate variability (HRV) ratio and conditioned pain modulation (CPM) response were measured and resting-state functional magnetic resonance imaging (MRI) was performed at baseline and after sublingual administration of either 0.2 mg/kg oral THC or placebo. RESULTS: THC significantly reduced the LF/HF ratio compared with placebo (interaction effect F(1,11) = 20.5; p < 0.005) and significantly improved CPM responses (interaction effect F(1,9) = 5.2; p = 0.048). The THC-induced reduction in LF/HF ratio correlated with increased functional connectivity between the rostral ventrolateral medulla and the dorsolateral prefrontal cortex [T(10) = 6.4, cluster p-FDR < 0.005]. CONCLUSIONS: THC shifts the autonomic balance towards increased parasympathetic tone and improves inhibitory pain mechanisms in chronic pain. The increase in vagal tone correlates with connectivity changes in higher-order regulatory brain regions, suggesting THC exerts top-down effects. These changes may reflect a normalizing effect of THC on multiple domains of supraspinal pain dysregulation. CLINICAL TRIAL REGISTRY NUMBER: NCT02560545.


Subject(s)
Chronic Pain , Neuralgia , Humans , Male , Dronabinol/pharmacology , Dronabinol/therapeutic use , Chronic Pain/drug therapy , Neuralgia/drug therapy , Brain , Double-Blind Method , Cross-Over Studies
8.
Maturitas ; 184: 107941, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38430618

ABSTRACT

INTRODUCTION: Medicinal cannabis might have a role in supporting the mental health of people with cancer. This systematic review and meta-analysis examined the efficacy and safety of medicinal cannabis, compared with any control, as an intervention for depression, anxiety, and stress symptoms in people living with cancer. A secondary aim was to examine the effect of low versus high Δ9-tetrahydrocannabinol (THC) dose on these outcomes. METHODS: Five databases were systematically searched, and complemented with a snowball search from inception to May 2023, for any type of interventional study that included humans of any age with any cancer type. Primary outcomes were incidence and severity of depression, anxiety, and stress symptoms. Secondary outcomes were mood, cognition, quality of life, appetite, nutrition status, gastrointestinal symptoms, and adverse events. Data were pooled using Review Manager. Evidence was appraised using Cochrane risk of bias tools. Confidence in the estimated effect of pooled outcomes was assessed using Grading of Recommendations, Assessment, Development and Evaluation (GRADE). RESULTS: Fifteen studies (n = 11 randomized trials, n = 4 non-randomized trials) of 18 interventions (N = 1898 total participants; 100 % ≥18 years of age) were included. Ten studies examined THC (70 % synthetic), two synthetic cannabidiol with or without THC, and six whole-plant extracts. No clinically significant effects of medicinal cannabis were found on primary outcomes. The likelihood of anxiety events increased with higher-dose synthetic THC compared with a lower dose (OR: 2.0; 95 % CI: 1.4, 2.9; p < 0.001; Confidence: very low). Medicinal cannabis (THC, cannabidiol, and whole-plant extract) increased the likelihood of improved appetite (OR: 12.3; 95 % CI: 3.5, 45.5; p < 0.001; n = 3 interventions; Confidence: moderate) and reduced severity of appetite loss (SMD: -0.4; 95 % CI: -0.8, -0.1; p = 0.009; Confidence: very low). There was very low confidence that higher doses of synthetic THC increased the likelihood of any adverse event (OR: 0.5; 95 % CI: 0.3, 0.7; p < 0.001). Medicinal cannabis had no effect on emotional functioning, mood changes, confusion, disorientation, quality of life, and gastrointestinal symptoms. Confidence in findings was limited by some studies having high or unclear risk of bias and imprecise pooled estimates. CONCLUSIONS: There was insufficient evidence to determine the efficacy and safety of medicinal cannabis as a therapeutic intervention for depression, anxiety, or stress in people with active cancer. Further research should explore whether medicinal cannabis might improve and maintain appetite and if high-dose synthetic THC might increase the incidence of side-effects, including anxiety. To inform clinical practice, well-powered and rigorously designed trials are warranted that evaluate the effects of medicinal cannabis prescribed to target anxiety, depression, and stress.


Subject(s)
Anxiety , Depression , Medical Marijuana , Neoplasms , Stress, Psychological , Humans , Neoplasms/drug therapy , Neoplasms/psychology , Medical Marijuana/therapeutic use , Medical Marijuana/adverse effects , Anxiety/drug therapy , Depression/drug therapy , Stress, Psychological/drug therapy , Dronabinol/pharmacology , Dronabinol/therapeutic use , Quality of Life
9.
Pharmacopsychiatry ; 57(3): 133-140, 2024 May.
Article in English | MEDLINE | ID: mdl-38471525

ABSTRACT

BACKGROUND: Up to now, it is unclear whether different medicinal cannabis (MC) strains are differently efficacious across different medical conditions. In this study, the effectiveness of different MC strains was compared depending on the disease to be treated. METHODS: This was an online survey conducted in Germany between June 2020 and August 2020. Patients were allowed to participate only if they received a cannabis-based treatment from pharmacies in the form of cannabis flowers prescribed by a physician. RESULTS: The survey was completed by n=1,028 participants. Most participants (58%) have used MC for more than 1 year, on average, 5.9 different strains. Bedrocan (pure tetrahydrocannabinol to pure cannabidiol [THC:CBD]=22:<1) was the most frequently prescribed strain, followed by Bakerstreet (THC:CBD=19:<1) and Pedanios 22/1 (THC:CBD=22:1). The most frequent conditions MC was prescribed for were different pain disorders, psychiatric and neurological diseases, and gastrointestinal symptoms. Overall, the mean patient-reported effectiveness was 80.1% (range, 0-100%). A regression model revealed no association between the patient-reported effectiveness and the variety. Furthermore, no influence of the disease on the choice of the MC strain was detected. On average, 2.1 side effects were reported (most commonly dry mouth (19.5%), increased appetite (17.1%), and tiredness (13.0%)). However, 29% of participants did not report any side effects. Only 398 participants (38.7%) indicated that costs for MC were covered by their health insurance. CONCLUSIONS: Patients self-reported very good efficacy and tolerability of MC. There was no evidence suggesting that specific MC strains are superior depending on the disease to be treated.


Subject(s)
Medical Marijuana , Humans , Germany , Male , Medical Marijuana/therapeutic use , Female , Adult , Middle Aged , Prospective Studies , Aged , Young Adult , Cannabidiol/therapeutic use , Surveys and Questionnaires , Adolescent , Dronabinol/therapeutic use , Cannabis , Treatment Outcome
10.
Pharmacopsychiatry ; 57(3): 160-168, 2024 May.
Article in English | MEDLINE | ID: mdl-38447959

ABSTRACT

Dementia, with loss of memory, cognitive abilities, and independent daily functioning, is increasing worldwide, related to an aging population. Currently, there is no curative treatment for dementia. Treatment of the frequently occurring behavioral and psychological symptoms of dementia (BPSD) is partially effective and associated with significant side effects. Cannabinoids are lipophilic molecules acting on the CB1 end CB2 receptors, essential for main biological processes such as sleep, appetite, memory, and pain. Cannabinoids might have a positive impact on amyloid formation in Alzheimer's disease, the main form of dementia, and on BPSD symptoms. Most knowledge currently concerns delta-9-tetrahydrocannabinol (THC) and cannabidiol (CBD). In the context of dementia and BPSD, THC might be beneficial for associated spasticity and possible pain or lack of appetite and CBD probably works better on sleep, agitation, and anxiety. This overview of prospective clinical studies and randomized clinical trials, published between 2005 and April 2023, using cannabinoids for BPSD suggests that older studies using low-dose oral synthetic THC showed no positive results. Still, more recent studies using THC/CBD-based oral medication at higher doses show promising results and are feasible and safe in this elderly polymedicated population. Several RCTs are ongoing and planned worldwide, and we hope other trials will follow to establish clinical efficiency and optimal dosing, as well as other outcomes such as deprescribing other medications and facilitation of care. We suggest that researchers also address the more sociological aspects of prescribing cannabinoids for dementia and BPSD in their specific context.


Subject(s)
Cannabinoids , Dementia , Humans , Dementia/drug therapy , Cannabinoids/therapeutic use , Behavioral Symptoms/drug therapy , Cannabidiol/therapeutic use , Dronabinol/therapeutic use
11.
Vet Immunol Immunopathol ; 269: 110727, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38330886

ABSTRACT

Dexamethasone (dex) is a potent glucocorticoid used to treat a variety of diseases. It is widely used in veterinary medicine in many species; for instance, in dogs, it can be used for emergent cases of anaphylaxis or trauma, management of immune-mediated hemolytic anemia or thrombocytopenia, certain cancers, allergic reactions, and topically for skin or eye inflammation. Dex is not without its side effects, especially when administered systemically, which might compromise compliance and effective treatment. Thus, adjunct therapies have been suggested to allow for decreased dex dosing and reduction in side effects while maintaining immunosuppressive efficacy. The goal of this study was to evaluate the potential for cannabinoids to serve as adjunct therapies for dex. Immune function was assessed in canine peripheral blood mononuclear cells (PBMCs) after treatment with dex with and without cannabidiol (CBD) and/or Δ9-tetrahydrocannabinol (THC). Dex suppressed IFN-γ protein secretion in a concentration-dependent manner and this suppression by low concentrations of dex was enhanced in the presence of CBD, THC, or the combination of CBD and THC. Similar effects were found with INFG and TNFA mRNA expression. These findings provide a rationale for using CBD or THC in vivo to reduce dex dosing and side effects.


Subject(s)
Cannabidiol , Cannabinoids , Dogs , Animals , Cannabinoids/therapeutic use , Dronabinol/therapeutic use , Leukocytes, Mononuclear , Cannabidiol/adverse effects , Dexamethasone/therapeutic use
12.
Mini Rev Med Chem ; 24(15): 1427-1448, 2024.
Article in English | MEDLINE | ID: mdl-38318827

ABSTRACT

Sativex is a cannabis-based medicine that comes in the form of an oromucosal spray. It contains equal amounts of Δ9-tetrahydrocannabinol and cannabidiol, two compounds derived from cannabis plants. Sativex has been shown to have positive effects on symptoms of amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS), and sleep disorders. It also has analgesic, antiinflammatory, antitumoral, and neuroprotective properties, which make it a potential treatment option for other neurological disorders. The article reviews the results of recent preclinical and clinical studies that support the therapeutic potential of Sativex and the molecular mechanisms behind its neuroprotective benefits in various neurological disorders. The article also discusses the possible advantages and disadvantages of using Sativex as a neurotherapeutic agent, such as its safety, efficacy, availability, and legal status.


Subject(s)
Cannabidiol , Dronabinol , Neuroprotective Agents , Plant Extracts , Humans , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemistry , Cannabidiol/pharmacology , Cannabidiol/therapeutic use , Cannabidiol/chemistry , Plant Extracts/chemistry , Plant Extracts/pharmacology , Dronabinol/pharmacology , Dronabinol/chemistry , Dronabinol/therapeutic use , Animals , Multiple Sclerosis/drug therapy , Cannabis/chemistry , Drug Combinations
13.
Aust N Z J Obstet Gynaecol ; 64(4): 411-415, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38415783

ABSTRACT

People with endometriosis use cannabis to manage their symptoms. This study aimed to identify costs, modes of administration, product composition, and self-reported effectiveness for those accessing medicinal cannabis in Australia. There were 192 survey responses analysed. Most (63.5%) used a 'cannabis clinic' doctor, incurring an initial consultation cost of $100-$200+ (10.2% Medicare bulk-billed) and median cannabinoid medicine costs of $300AUD per month. Cost was a major barrier to access, necessitating reducing dosage (76.1%) and/or consuming illicit cannabis (42.9%), despite a prescription. Most (77%) medical consumers used two or more cannabis products, with delta-9-tetrahydrocannabinol predominant oil and flower products most frequently prescribed.


Subject(s)
Cannabinoids , Endometriosis , Medical Marijuana , Humans , Female , Endometriosis/drug therapy , Endometriosis/economics , Australia , Medical Marijuana/therapeutic use , Cannabinoids/therapeutic use , Cannabinoids/economics , Adult , Health Services Accessibility , Surveys and Questionnaires , Middle Aged , Young Adult , Dronabinol/therapeutic use , Australasian People
14.
Neurodegener Dis Manag ; 14(1): 11-20, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38318862

ABSTRACT

Aim: This prospective, multicenter, open-label, noninterventional 12-week study investigated the effectiveness and tolerability of add-on nabiximols oromucosal spray (Sativex®) in the real-world setting in Germany. Patients & methods: The main analysis set comprised 51 adult patients (49 nabiximols responders) with multiple sclerosis (MS) spasticity. Results: The mean overall goal attainment scale score (primary outcome measure) increased by 46% from baseline to week 12 (35.2 vs 51.4; p < 0.001). Mean gait speed was improved by 23% at 4 and 12 weeks. Clinically meaningful improvements in mean 0-10 numerical rating scale scores for spasticity, pain, sleep quality and urinary bladder dysfunction were recorded at 4 and 12 weeks. Conclusion: Nabiximols is a useful therapeutic option for patients with MS spasticity.


People with multiple sclerosis (MS) spasticity experience a variety of symptoms and have individual expectations about a new treatment. This study investigated patients' perceptions about the effectiveness and tolerability of nabiximols oromucosal spray (Sativex®) when added to current medications for spasticity. Common treatment goals for patients (n = 51) were less pain, better walking and improved sleep. After 12 weeks of treatment, 62% of selected treatment goals were achieved 'as expected' or 'better than expected' and 65% of patients considered their spasticity to be 'much improved'. Meaningful improvements were recorded in spasticity-related symptoms of pain, sleep quality and bladder problems. Few side effects were reported. Nabiximols may be useful for MS patients with a poor response to usual spasticity medications.


Subject(s)
Cannabidiol , Multiple Sclerosis , Adult , Humans , Cannabidiol/therapeutic use , Dronabinol/therapeutic use , Drug Combinations , Multiple Sclerosis/complications , Multiple Sclerosis/drug therapy , Muscle Spasticity/drug therapy , Muscle Spasticity/etiology , Patient Reported Outcome Measures , Plant Extracts/therapeutic use , Prospective Studies , Treatment Outcome
15.
Pain Manag Nurs ; 25(1): e8-e20, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37689509

ABSTRACT

OBJECTIVE: To assess cannabinoid dosing that could be associated with a reduction in opioid use. DESIGN: Systematic review conducted according to the PRISMA statement. DATA SOURCES: PubMed, Embase, Web of Science, and PsycINFO were searched up to December 10, 2022. REVIEW/ANALYSIS METHODS: We included randomized controlled trials (RCT) and longitudinal observational studies assessing cannabinoids effect on opioid use in patients with acute or chronic pain. Two reviewers independently assessed the studies for inclusion and extracted the data. Tetrahydrocannabinol (THC), Cannabidiol (CBD), and other cannabinoids with dosing were the exposures. Change in opioid doses and opioid discontinuation were the outcomes. RESULTS: Fifteen studies (including seven RCTs) were included. Eight studies (six observational and two RCTs) were conducted among patients with chronic pain including three with cancer-related pain. Seven studies involved patients with acute pain (five RCTs).In chronic non-cancer pain patients, two observational studies that assessed THC and CBD in combination (average daily dose 17mg/15mg), and one that assessed a CBD-rich extract (31.4 mg/day), showed a significant reduction in opioid use. Of the three studies conducted on patients with cancer, only the observational study that assessed nabilone (average 1.7 mg/day) showed a significant reduction in opioid use. In patients with acute pain, only two observational studies that assessed dronabinol (5mg and 5-10 mg/day for four days) showed a significant reduction in opioid use. CONCLUSION: The opioid-sparing effect of cannabinoids remains uncertain based on current evidence. However, attention could be paid to cannabinoid doses associated with opioid reduction in included observational studies.


Subject(s)
Acute Pain , Cannabinoids , Chronic Pain , Opiate Substitution Treatment , Humans , Acute Pain/drug therapy , Analgesics, Opioid/therapeutic use , Cannabidiol/adverse effects , Cannabinoids/therapeutic use , Chronic Pain/drug therapy , Dronabinol/therapeutic use , Longitudinal Studies , Observational Studies as Topic
16.
Curr Pain Headache Rep ; 28(3): 109-118, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38095748

ABSTRACT

PURPOSE OF REVIEW: Neuropathic pain (NP) remains a challenge to treat, with 50% of patients experiencing limited efficacy from current treatments. Medicinal cannabis, which contains tetrahydrocannabinol (THC), cannabidiol (CBD) and other minor cannabinoids, is garnering attention as an alternative treatment for NP. This paper reviews the clinical evidence for phytocannabinoid treatment of NP. RECENT FINDINGS: Seventeen randomised controlled trials (RCT) were identified for inclusion in this review. Of these, ten studies using phytocannabinoid preparations containing THC alone had the most evidence for pain relief. Four studies investigating THC/CBD combinations showed some reductions in pain scores, although not all findings were statistically significant, whereas studies investigating CBD (two studies) or cannabidivarin (one study) showed no analgesic effect over placebo. However, CBD studies were of small sample size when compared to other studies in the review and short duration. Results for treatment of diabetic peripheral neuropathy patients with THC showed better improvements over those for NP induced by chemotherapy and multiple sclerosis, with these trials using vaporised whole plant cannabis. This formulation may have trace amounts of other minor cannabinoids, compared with synthetic cannabinoids such as dronabinol or nabilone that were investigated in other studies. This review provides an overview of RCTs that have investigated phytocannabinoid use for the treatment of NP. There appears to be evidence to necessitate further high quality RCTs into novel formulations of phytocannabinoids for the treatment of NP.


Subject(s)
Cannabinoids , Cannabis , Medical Marijuana , Neuralgia , Humans , Dronabinol/therapeutic use , Dronabinol/pharmacology , Cannabinoids/therapeutic use , Neuralgia/drug therapy , Medical Marijuana/therapeutic use , Randomized Controlled Trials as Topic
17.
Chin J Physiol ; 66(6): 446-455, 2023.
Article in English | MEDLINE | ID: mdl-38149557

ABSTRACT

Despite the current optimal therapy, patients with myocardial ischemia/reperfusion (IR) injury still experience a high mortality rate, especially when diabetes mellitus is present as a comorbidity. Investigating potential treatments aimed at improving the outcomes of myocardial IR injury in diabetic patients is necessary. Our objective was to ascertain the cardioprotective effect of delta 9-tetrahydrocannabinol (THC) against myocardial IR injury in diabetic rats and examine the role of phosphatase and tensin homolog (PTEN)/phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) signaling pathway in mediating this effect. Diabetes was induced in male Wistar rats (8-10 weeks old, 200-250 g; n = 60) by a single injection of streptozotocin. The duration of the diabetic period was 10 weeks. During the last 4 weeks of diabetic period, rats were treated with THC (1.5 mg/kg/day; intraperitoneally), either alone or in combination with LY294002, and then underwent IR intervention. After 24 h of reperfusion, infarct size, cardiac function, lactate dehydrogenase (LDH) and cardiac-specific isoform of troponin-I (cTn-I) levels, myocardial apoptosis, oxidative stress markers, and expression of PTEN, PI3K, and Akt proteins were evaluated. THC pretreatment resulted in significant improvements in infarct size and cardiac function and decreases in LDH and cTn-I levels (P < 0.05). It also reduced myocardial apoptosis and oxidative stress, accompanied by the downregulation of PTEN expression and activation of the PI3K/Akt signaling pathway (P < 0.05). LY294002 pretreatment abolished the cardioprotective action of THC. This study revealed the cardioprotective effects of THC against IR-induced myocardial injury in diabetic rats and also suggested that the mechanism may be associated with enhanced activity of the PI3K/Akt signaling pathway through the reduction of PTEN phosphorylation.


Subject(s)
Diabetes Mellitus, Experimental , Myocardial Reperfusion Injury , Humans , Rats , Male , Animals , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/pharmacology , Phosphatidylinositol 3-Kinase/metabolism , Phosphatidylinositol 3-Kinase/pharmacology , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/pharmacology , Dronabinol/pharmacology , Dronabinol/therapeutic use , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Rats, Sprague-Dawley , Rats, Wistar , Signal Transduction , Infarction , Apoptosis , PTEN Phosphohydrolase/metabolism , PTEN Phosphohydrolase/pharmacology
18.
JBJS Rev ; 11(11)2023 Nov 01.
Article in English | MEDLINE | ID: mdl-37972215

ABSTRACT

¼ Cannabinoids, such as D9-tetrahydrocannabinol and cannabidiol, interact with endocannabinoid receptors in the central nervous system and immune system, potentially offering pain relief. The entourage effect, resulting from the interaction of multiple cannabis components, may enhance therapeutic impact and efficacy, making them promising candidates for exploring pain relief in spine operations, known to be among the most painful operative procedures.¼ The use of cannabinoids in pain management requires careful consideration of safety, including their cognitive and psychomotor effects, potential cardiovascular risks, risk of dependence, mental health implications, and drug interactions.¼ Few studies have analyzed cannabinoid use in relation to spine surgery, with variable results reported, indicating possible effects on reoperation rates, mortality, complications, postoperative opioid use, and length of hospital stay.¼ Current knowledge gaps exist in the understanding of cannabinoid effects on spine surgery, including the exploration of different administration routes, timing, dosage, and specific outcomes. In addition, mechanistic explanations for the observed results are lacking.¼ Ethical considerations related to informed consent, medical expertise, societal impact, and legal compliance must also be thoroughly addressed when considering the utilization of cannabinoids in spinal pathologies and back pain treatment.


Subject(s)
Cannabinoids , Cannabis , Humans , Cannabinoids/therapeutic use , Pain Management , Dronabinol/therapeutic use , Pain
19.
CNS Drugs ; 37(11): 981-992, 2023 11.
Article in English | MEDLINE | ID: mdl-37945917

ABSTRACT

BACKGROUND AND OBJECTIVES: Medical cannabis use is increasing in Australia and other jurisdictions, yet little is known about the effects of medical cannabis on cognitive function. Findings from studies of non-medical ('recreational') cannabis may not be applicable to patients using prescribed medical cannabis to manage a health condition. METHODS: In this semi-naturalistic, open-label trial, patients with various health conditions attended a single laboratory session in which they self-administered a standard dose of prescribed medical cannabis as per instructions on the pharmacy label. We assessed cognitive performance using the Cambridge Neuropsychological Test Automated Battery (CANTAB) and Druid application (app) prior to and following (CANTAB: + 3 h; Druid: + 3 and 5.5 h) medical cannabis self-administration. We also assessed subjective drug effects prior to and following (1, 2 and 4 h) medical cannabis self-administration using a range of 0-10 cm visual analogue scales ('stoned', 'sedated', 'relaxed', 'comfortable', 'anxious' and 'confident'). Data were analyzed using linear fixed-effect models. RESULTS: Participants (N = 40; 22 females) were prescribed a range of products including orally administered oils (n = 23) and flower for vaporization (n = 17). Participants had a mean (standard deviation [SD]) age of 41.38 (12.66) years and had been using medical cannabis for a mean (SD) of 10.18 (8.73) months. Chronic non-cancer pain was the most common indication for medical cannabis use (n = 20), followed by sleep disorder (n = 18) and anxiety (n = 11). The mean (SD) delta-9-tetrahydrocannabinol (THC)/cannabidiol (CBD) dose administered by participants was 9.61 (8.52) mg/9.15 (10.11) mg among those using an oil, and 37.00 (24.53) mg/0.38 (1.58) mg among those who vaporized flower, respectively. Participants' performance improved over time on the CANTAB Multitasking Test and Rapid Visual Information Processing test (both p-values <0.001). All other changes in cognitive performance measures over time were non-significant (p > 0.05). Vaporization of flower was associated with significantly stronger subjective feelings of 'stoned' and 'sedated' relative to oils (both p < 0.001). CONCLUSIONS: These findings suggest that prescribed medical cannabis may have minimal acute impact on cognitive function among patients with chronic health conditions, although larger and controlled trials are needed.


Subject(s)
Chronic Pain , Marijuana Smoking , Medical Marijuana , Adult , Female , Humans , Analgesics, Opioid , Dronabinol/pharmacology , Dronabinol/therapeutic use , Marijuana Smoking/psychology , Medical Marijuana/adverse effects , Oils
20.
Einstein (Sao Paulo) ; 21: eRW0387, 2023.
Article in English | MEDLINE | ID: mdl-37991091

ABSTRACT

INTRODUCTION: The increasing popularity of cannabinoids for treating numerous neurological disorders has been reported in various countries. Although it reduces tetrahydrocannabinol psychoactivity, it helps patients tolerate higher doses and complements the anti-spasmodic effects of tetrahydrocannabinol. One of the most important potential of cannabinoids are related to its potential to help children with cerebral palsy, a contributor of lifelong disability. Therefore, this systematic review aimed to assess the efficacy and safety of medical cannabinoids in children with cerebral palsy. METHODS: This review adhered to The Preferred Reporting Items for Systematic Reviews and Meta-analysis 2020 guidelines. Seven databases, namely, Scopus, PubMed, EBSCO Host, ProQuest, Google Scholar, Semantic Scholar, and JSTOR, were used to identify relevant studies. Studies examining pediatric patients with cerebral palsy and reporting the efficacy and safety of medical cannabinoids through clinical trials, observational cross-sectional studies, or cohort designs were included. The outcomes of the studies included the efficacy of medical cannabinoids administered for spasticity, motor components, pain control, sleep difficulties, adverse effects, and seizure control. RESULTS: Of 803 identified articles, only three met the inclusion criteria for data synthesis. One study exhibited a moderate risk-of-bias. A total of 133 respondents, mainly from Europe, were investigated. Overall effectiveness and safety were considered good. However, the results are inconsistent, especially regarding spasticity treatment variables. CONCLUSION: The anti-spasticity, anti-inflammatory, and anti-seizure properties of cannabinoids might be beneficial for patients with cerebral palsy, although their effectiveness has not been widely studied. Further studies with larger sample sizes and various ethnicities are warranted. Prospero database registration: (www.crd.york.ac.uk/prospero) under ID CRD42022358383.


Subject(s)
Cannabinoids , Cerebral Palsy , Child , Humans , Cerebral Palsy/drug therapy , Cannabinoids/adverse effects , Dronabinol/therapeutic use , Cross-Sectional Studies , Muscle Spasticity/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL