Acessibilidade / Reportar erro

Medical cannabinoids for treatment of neuropsychiatric symptoms in dementia: a systematic review

Abstract

Introduction

Neuropsychiatric symptoms are an integral component of the natural history of dementia, occurring from prodromal to advanced stages of the disease process and causing increased burden and morbidity. Clinical presentations are pleomorphic and clinical management often requires combinations of pharmacological and non-pharmacological interventions. However, limited efficacy and a non-negligible incidence of adverse psychotropic drug events emphasize the need for novel therapeutic options.

Objectives

To review the evidence supporting use of medical cannabinoids for treatment of neuropsychiatric symptoms (NPS) of dementia.

Methods

We conducted a systematic review of the medical literature to examine scientific publications reporting use of medical cannabinoids for treatment of NPS. Medical Subject Headings (MeSH) were used to search for relevant publications and only papers reporting original clinical information were included. A secondary search was performed within selected publications to capture relevant citations that were not retrieved by the systematic review. The papers selected were categorized according to the level of evidence generated by the studies in relation to this clinical application, i.e. (1) controlled clinical trials; (2) open-label or observational studies; and (3) case reports.

Results

Fifteen publications with original clinical data were retrieved: five controlled clinical trials, three open-label/observational studies, and seven case reports. Most studies indicated that use of medical cannabinoids engendered favorable outcomes for treatment of NPS related to moderate and advanced stages of dementia, particularly agitation, aggressive behavior, sleep disorder, and sexual disinhibition.

Conclusion

Medical cannabinoids constitute a promising pharmacological approach to treatment of NPS with preliminary evidence of benefit in at least moderate to severe dementia. Controlled trials with longitudinal designs and larger samples are required to examine the long-term efficacy of these drugs in different types and stages of dementia, in addition to their adverse events and risk of interactions with other drugs. Many pharmacological details are yet to be determined, such as dosing, treatment duration, and concentrations of active compounds (e.g., cannabidiol [CBD]/ Δ9-tetrahydrocannabinol [THC] ratio) in commercial preparations of medical cannabinoids.

Dementia; neuropsychiatric symptoms; cannabidiol; Δ9-tetrahydrocannabinol; treatment

Introduction

Neuropsychiatric symptoms (NPS) are relevant clinical manifestations that pertain to the natural history of dementia, with prevalence estimates ranging from 35 to 95% depending on the severity of clinical deterioration.11. Steinberg M, Shao H, Zandi P, Lyketsos CG, Welsh-Bohmer KA, Norton MC, et al. Point and 5-year period prevalence of neuropsychiatric symptoms in dementia: the Cache County Study. Int J Geriatr Psychiatry. 2008;23:170-77.

2. Lyketsos CG. Neuropsychiatric symptoms in dementia: overview and measurement challenges. J Prev Alzheimers Dis. 2015;2:155-6.
- 33. Radue R, Walaszek A, Asthana S. Neuropsychiatric symptoms in dementia. Handb Clin Neurol. 2019;167:437-54. These manifestations are also referred to as behavioral and psychological symptoms of dementia (BPSD), and may present with severe and disabling behaviors, critically impacting on the well-being of both patients and caregivers. NPS are ubiquitous across different etiologies of dementia disorders, but particularly important in Alzheimer’s disease (AD), frontotemporal dementia (FTD), Lewy body dementia (LBD), and other forms of dementia with Parkinsonism. In AD, for instance, NPS can be observed from the pre-dementia phase through all stages of dementia, with increasing incidence and magnitude as the disease progresses.44. Jost BC, Grossberg GT. The evolution of psychiatric symptoms in Alzheimer’s disease: a natural history study. J Am Geriatr Society. 1996;44:1078-81. , 55. Mega MS, Cummings JL, Fiorello T, Gornbein J. The spectrum of behavioral changes in Alzheimer’s. Neurology. 1996;46:130-5. At prodromal and preclinical stages of AD, the occurrence of NPS is associated with increased risk of conversion to dementia and subsequent cognitive and functional deterioration.66 Stella F, Radanovic M, Balthazar MLF, Canineu PR, de Souza LC, Forlenza OV. Neuropsychiatric symptoms in the prodromal stages of dementia. Curr Opin Psychiatry.2014;27:230-5. In most etiologies, NPS tend to become more prevalent with progression of the neurodegenerative processes, in which case symptoms and abnormal behaviors may illustrate the damage to specific brain areas.22. Lyketsos CG. Neuropsychiatric symptoms in dementia: overview and measurement challenges. J Prev Alzheimers Dis. 2015;2:155-6. , 33. Radue R, Walaszek A, Asthana S. Neuropsychiatric symptoms in dementia. Handb Clin Neurol. 2019;167:437-54. Relevant and persistent behavioral abnormalities have been related to increased morbidity and higher mortality risk. As a general rule, the occurrence of persistent symptoms often accounts for debilitating outcomes and predicts more severe global deterioration.22. Lyketsos CG. Neuropsychiatric symptoms in dementia: overview and measurement challenges. J Prev Alzheimers Dis. 2015;2:155-6. Also, NPS invariably increase caregiver burden, being associated with early nursing home placement and hastening the decision to institutionalize.77. Boccardi V, Conestabile Della Staffa M, Baroni M, Ercolani S, Croce MF, Ruggiero C, et al. Prevalence and correlates of behavioral disorders in old age subjects with cognitive impairment: results from the ReGAl project. J Alzheimers Dis. 2017;60:1275-83.

Effective treatment of NPS is still a challenge in clinical practice. At earlier stages of dementia in AD and related disorders, regular use of cholinesterase inhibitors and meantime in therapeutic doses is recommended as the first step in the pharmacological management of mild behavioral changes. However, in the presence of severe and disruptive behaviors such as psychosis and agitation, which often occur in advanced stages of dementia, other psychotropic drugs must be considered, including antipsychotics, antidepressants, benzodiazepines, and anticonvulsants. Nonetheless, studies of psychopharmacological interventions for treatment of NPS have so far yielded at best modest evidence of efficacy, which must be counterbalanced by relevant safety concerns. Special attention must be given to increased risk of cerebrovascular events and all-cause mortality, especially when these medications are used for extended periods.88. Maust DT, Kim HM, Seyfried LS, Chiang C, Kavanagh J, Schneider LS, et al. Antipsychotics, other psychotropics, and the risk of death in patients with dementia: number needed to harm. JAMA Psychiatry. 2015;72:438-45.

9. Kales HC, Lyketsos CG, Miller EM, Ballard C. Management of behavioral and psychological symptoms in people with Alzheimer’s disease: an international Delphi consensus. Int Psychogeriatr. 2019;31:83-90.
- 1010. Mueller C, John C, Perera G, Aarsland D, Ballard C, Stewart R. Antipsychotic use in dementia: the relationship between neuropsychiatric symptom profiles and adverse outcomes. Eur J Epidemiol. 2021;36:89-101. On the other hand, non-pharmacological strategies, although more acceptable and safer, may not be efficacious (or even inapplicable) in certain types of NPS, particularly the most severe. Nonetheless, it is widely accepted that these interventions must accompany and if possible precede introduction of psychotropic drugs for clinical management of NPS, with recognizable benefits for general state of health.1111. Brodaty H, Arasaratnam C. Meta-analysis of nonpharmacological interventions for neuropsychiatric symptoms of dementia. Am J Psychiatry. 2012;169:946-53. In this context, new therapeutic options to treat NPS are needed and eagerly awaited by the clinical and scientific community.

The endocannabinoid system (eCB) plays important homeostatic roles, modulating multiple physiological functions through signaling from widespread receptors (CB1 and CB2) present in the peripheral and central nervous systems. Systemically, the eCB is relevant to cardiovascular, immunological, and reproductive functions.1212. Fernández-Ruiz J, Pazos MR, García-Arencibia M, Sagredo O, Ramos JA. Role of CB2 receptors in neuroprotective effects of cannabinoids. Mol Cell Endocrinol. 2008;286:S91-6. In the brain, with signaling predominantly through CB1 receptors, it impacts on distinct neurobehavioral functions that range from cognition (learning and memory) to behavior (mood regulation, emotional control, appetite, feeding, and addictive behavior), in addition to sensitivity to pain and mechanisms of neuroprotection.1313. Busquets-Garcia A, Bains J, Marsicano G. CB1 Receptor signaling in the brain: extracting specificity from ubiquity. Neuropsychopharmacology. 2018;43:4-20. Expression of CB1 receptors in the basal ganglia regulates locomotor activity,1414. Navarro G, Morales P, Rodríguez-Cueto C, Fernández-Ruiz J, Jagerovic N, Franco R. Targeting cannabinoid CB2 receptors in the central nervous system. medicinal chemistry approaches with focus on neurodegenerative disorders. Front Neurosci. 2016;10:406. whereas their expression in the cortex and hippocampus is predominantly related to cognitive-behavioral effects as well as to psychotropic and antiepileptic properties. The ubiquitous expression of cannabinoid receptors in the brain also illustrates their potential to interact with other neurotransmitter systems, such as those mediated by acetylcholine, dopamine, GABA, histamine, serotonin, glutamate, norepinephrine, prostaglandins, and opioid peptides, explaining the pleomorphic pharmacological effects of cannabinoid drugs.

Pharmacological compounds that modulate the eCB have been more extensively tested in certain neurological and medical conditions, such as Parkinson’s disease, epilepsy, chronic pain, and incoercible nausea/vomiting secondary to cancer treatments.1515. Burggren AC, Shirazi A, Ginder N, London ED. Cannabis effects on brain structure, function, and cognition: considerations for medical uses of cannabis and its derivatives. Am J Drug Alcohol Abuse. 2019;45:563-79. Expert reviews further suggest that use of these drugs may also be justified for treatment of autism, bipolar disorder, schizophrenia, Tourette syndrome, and depression, although construction of scientific evidence to support such indications is still in progress.1515. Burggren AC, Shirazi A, Ginder N, London ED. Cannabis effects on brain structure, function, and cognition: considerations for medical uses of cannabis and its derivatives. Am J Drug Alcohol Abuse. 2019;45:563-79. Use of medical cannabinoids for management of NPS has attracted the attention of both clinicians and researchers in the recent past, envisaging the potential benefits of cannabidiol (CBD), Δ99. Kales HC, Lyketsos CG, Miller EM, Ballard C. Management of behavioral and psychological symptoms in people with Alzheimer’s disease: an international Delphi consensus. Int Psychogeriatr. 2019;31:83-90. -tetrahydrocannabinol (THC), and combined preparations for management of difficult clinical situations. Preliminary and uncontrolled communications reported positive effects on severe and refractory symptoms that were otherwise unmanageable, particularly in cases of advanced dementia.1616. Passmore MJ. The cannabinoid receptor agonist nabilone for the treatment of dementia-related agitation. Int J Geriatr Psychiatry. 2008;23:116-7. However, the body of evidence to support use and to guide the prescription of medical cannabinoids for the treatment of NPS is still controversial. Therefore, the objective of the present study is to systematically review the medical literature to examine studies reporting on use of medical cannabinoids for treatment of NPS and to compile the available data on efficacy, tolerability, and dose regimens.

Methods

We conducted a systematic and critical analysis of the specialized literature, searching for articles containing original data published in English in the PubMed database, using Medical Subject Headings (MeSH) terms. Thus, the following inclusion criteria guided the search strategy: (cannabinoid* or cannabidiol or CBD or Δ9-tetrahydrocannabinol or THC or endocannabinoid* or marijuana or nabilone or dronabinol) and (dementia or “cognitive impairment” or “cognitive decline” or Alzheimer* or “vascular dementia” or “frontotemporal dementia” or “semantic dementia” or aphasia or “language dementia” or “Lewy bodies” or “dementia in psychiatric diseases”) and (“behavioral and psychological symptoms of dementia” or BPSD or “neuropsychiatric symptoms” or agitation or aggression or delusions or hallucinations or depression or anxiety or irritability or apathy or “sleep disorders” or insomnia or “appetite disorder” or disinhibition or cognition or cognitive). We also performed a manual search for relevant citations identified in the selected articles that were not retrieved by the systematic search. Exclusion criteria comprised studies in which psychopathological manifestations were not the main target of treatment with medical cannabinoids, and those whose NPS did not arise from a dementia condition or were not clearly defined. To identify additional references that may have been missed by the systematic review, we conducted an active search of the citations from selected articles. Review articles, editorials, points of view, study protocols, and secondary analyses were ruled out, i.e., only papers reporting original clinical information were included in the analysis. We established three categories to rank publications according to the respective level of evidence provided on the efficacy of these compounds for treatment of NPS: controlled clinical trials with standardized NPS assessment (evidence level 1); open-label or observational studies with standardized NPS assessment (level 2); and case reports with or without standardized NPS assessment (level 3). The search covered a period extending from the 1990s to 28th February, 2021. Two researchers extracted the data from the reports (FS and LCLV).

We further analyzed the data according to clusters of target symptoms: agitation/aggression (including wandering, erratic motor behavior, threatening behavior, aggressive behavior); sleep disturbances (insomnia, nighttime behavior); and abnormal sexual behavior (sexual disinhibition, inappropriate sexual comments, public masturbation, touching).

Results

The search strategy yielded a total of 801 publications. Most of these did not meet the inclusion criteria, resulting in only 15 studies that reported original, clinical data ( Figure 1 ). Of these, five studies were controlled trials1717. Volicer L, Stelly M, Morris J, McLaughlin J, Volicer BJ. Effects of dronabinol on anorexia and disturbed behavior in patients with Alzheimer’s disease. Int J Geriatr Psychiatry. 1997;12:913-9.

18. Mahlberg R, Walther S. Actigraphy in agitated patients with dementia - actigraphy in agitated patients with dementia: monitoring treatment outcomes. Z Gerontol Geriatr. 2007;40:178-84.

19. van den Elsen GAH, Ahmed AIA, Verkes R-J, Feuth T, van der Marck MA, Olde Rikkert MGM. Tetrahydrocannabinol in behavioral disturbances in dementia: a crossover randomized controlled trial. Am J Geriatr Psychiatry. 2015;23:1214-24a.

20. van den Elsen GAH, Ahmed AIA, Verkes R-J, Kramers C, Feuth T, Rosenberg PB, et al. Tetrahydrocannabinol for neuropsychiatric symptoms in dementia: a randomized controlled trial. Neurology. 2015;84:2338-46b.
- 2121. Herrmann N, Ruthirakuhan M, Gallagher D, Verhoeff NPLG, Kiss A, Black SE, et al. Randomized placebo-controlled trial of nabilone for agitation in Alzheimer’s disease. Am J Geriatr Psychiatry. 2019;27:1161-73. , three were open-label/observational studies, and seven were case reports.1616. Passmore MJ. The cannabinoid receptor agonist nabilone for the treatment of dementia-related agitation. Int J Geriatr Psychiatry. 2008;23:116-7. , 2222. Walther S, Mahlberg R, Eichmann U, Kunz D. Delta-9-tetrahydrocannabinol for nighttime agitation in severe dementia. Psychopharmacology. 2006;185:524-8.

23. Shelef A, Barak Y, Berger U, Paleacu D, Tadger S, Plopsky I, et al. Safety and efficacy of medical cannabis oil for behavioral and psychological symptoms of dementia: an-open label, add-on, pilot study. J Alzheimers Dis. 2016;51:15-9.

24. Broers B, Patà Z, Mina A, Wampfler J, de Saussure C, Pautex S. Prescription of a THC/CBD-Based medication to patients with dementia: a pilot study in Geneva. Med Cannabis Cannabinoids. 2019;2:56-9.

25. Walther S, Schüpbach B, Seifritz E, Homan P, Strik W. Randomized, controlled crossover trial of dronabinol, 2.5 mg, for agitation in 2 patients with dementia. J Clin Psychopharmacol. 2011;31:256-8.

26. Amanullah S, MacDougall K, Sweeney N, Coffin J, Cole J. Synthetic cannabinoids in dementia with agitation: case studies and literature review. Clin Neuropsychiatry. 2013;10:142-7.

27. Zajac DM, Sikkema SR, Chandrasena R. Nabilone for the treatment of dementia-associated sexual disinhibition. Prim Care Companion CNS Disord. 2015;17: 10.4088/PCC.14l01695.
https://doi.org/10.4088/PCC.14l01695...

28. Wilhelm R, Ahl B, Anghelescu I-G. Dextrometorphan-paroxetine, but not dronabinol, effective for treatment-resistant aggression and agitation in an elderly patient with lewy body dementia. J Clin Psychopharmacol. 2017;37:745-7.

29. Defrancesco M, Hofer A. Cannabinoid as beneficial replacement therapy for psychotropics to treat neuropsychiatric symptoms in severe Alzheimer’s dementia: a clinical case report. Front Psychiatry. 2020;11:413.
- 3030. Gopalakrishna G, Srivathsal Y, Kaur G. Cannabinoids in the management of frontotemporal dementia: a case series. Neurodegener Dis Manag. 2021;11:61-4. To facilitate description of the findings, the publications selected were grouped in the following subsections, according to the neuropsychiatric domains targeted by the intervention: (a) agitation; (b) sleep disturbances; (c) abnormal sexual behavior.

Figure 1
Schematic flow diagram illustrating literature search, articles excluded, and articles included in the review.

Agitation

Controlled clinical trials

In a placebo-controlled study with a crossover design, Volicer et al.1717. Volicer L, Stelly M, Morris J, McLaughlin J, Volicer BJ. Effects of dronabinol on anorexia and disturbed behavior in patients with Alzheimer’s disease. Int J Geriatr Psychiatry. 1997;12:913-9. investigated the effect of dronabinol (an orexigenic THC formulation) on a fixed dose schedule (2.5 mg/day) for management of food refusal and behavioral disturbances in 15 patients with AD, most suffering from severe dementia.1515. Burggren AC, Shirazi A, Ginder N, London ED. Cannabis effects on brain structure, function, and cognition: considerations for medical uses of cannabis and its derivatives. Am J Drug Alcohol Abuse. 2019;45:563-79. Dronabinol was prescribed for 6 weeks, followed by placebo for a similar period. Eleven patients completed the study. Treatment with dronabinol substantially improved anorexia and weight gain, and also reduced the severity of behavioral disorders, e.g., agitation and euphoria. Somnolence and tiredness were the most commonly reported adverse reactions, although not requiring the intervention to be discontinued. However, one patient developed a generalized seizure, an adverse event that was regarded as possibly related to the drug.

Mahlberg et al.1818. Mahlberg R, Walther S. Actigraphy in agitated patients with dementia - actigraphy in agitated patients with dementia: monitoring treatment outcomes. Z Gerontol Geriatr. 2007;40:178-84. examined two pharmacological compounds for treatment of patients with severe dementia due to AD (mean age 79.0 years) suffering from agitation, specifically nocturnal behavior. The authors prescribed dronabinol 2.5 mg daily for seven patients and melatonin 3.0 mg per day for another seven patients and compared these therapies over a 2-week period to 10 patients of similar age who received placebo. Nocturnal behavior was measured by means of actigraphy. Patients in both intervention groups (dronabinol or melatonin) showed a significant improvement in nocturnal agitation as compared to patients in the placebo group. However, an important methodological limitation concerns the aggregation of dronabinol and melatonin patients in the same group, limiting the assessment of the efficacy of dronabinol. Safety and tolerability were not described, which constitutes another limitation.

In a small randomized, controlled trial, van den Elsen et al.1919. van den Elsen GAH, Ahmed AIA, Verkes R-J, Feuth T, van der Marck MA, Olde Rikkert MGM. Tetrahydrocannabinol in behavioral disturbances in dementia: a crossover randomized controlled trial. Am J Geriatr Psychiatry. 2015;23:1214-24a. evaluated the effect of low doses of oral THC for treatment of neuropsychiatric manifestations for 12 weeks in a test group of 22 patients (mean age 76.4 years) with AD, vascular dementia (VD), or mixed dementia (AD/VD) presenting with severe NPS.1919. van den Elsen GAH, Ahmed AIA, Verkes R-J, Feuth T, van der Marck MA, Olde Rikkert MGM. Tetrahydrocannabinol in behavioral disturbances in dementia: a crossover randomized controlled trial. Am J Geriatr Psychiatry. 2015;23:1214-24a. Initially, patients received a low THC dosage of 0.75 mg twice daily, which was subsequently shifted to placebo. Thereafter, the THC dosage was increased to 1.5 mg twice a day and, again, was substituted with placebo. There were no advantages of THC compared to placebo in the measurement of NPS, including agitation and aggression. Nonetheless, low-dose THC was well tolerated and safe, supporting additional studies with escalating doses.

The same group conducted a randomized, double-blind, placebo-controlled trial to examine the efficacy and safety of low-dose oral THC (4.5 mg/day) for 3 weeks for treatment of NPS in patients with severe AD, VD, or AD/VD.2020. van den Elsen GAH, Ahmed AIA, Verkes R-J, Kramers C, Feuth T, Rosenberg PB, et al. Tetrahydrocannabinol for neuropsychiatric symptoms in dementia: a randomized controlled trial. Neurology. 2015;84:2338-46b. The intervention group (n = 24, mean age 79.0 years) received dronabinol (1.5 mg t.i.d. [ ter in die , three times a day]), while the comparison group (n = 26, mean age 78.0 years) received placebo. Primary outcomes were negative, given that treatment with dronabinol did not reduce NPS in this study group. Nevertheless, the THC compound was well tolerated and patients had no clinically relevant side effects on vital signs, weight, or episodic memory.

Herrmann et al.2121. Herrmann N, Ruthirakuhan M, Gallagher D, Verhoeff NPLG, Kiss A, Black SE, et al. Randomized placebo-controlled trial of nabilone for agitation in Alzheimer’s disease. Am J Geriatr Psychiatry. 2019;27:1161-73. conducted a randomized, double-blind, crossover trial with nabilone, a THC-analogue, synthetic cannabinoid that is used therapeutically for its antiemetic, orexigenic, and analgesic properties. Thirty-eight patients with moderate and severe AD (mean age 87.0 years) and treatment-resistant agitation were given 1.0-2.0 mg of nabilone daily for 14 weeks. The first part of the intervention consisted of a 1-week placebo phase followed by 6 weeks of nabilone treatment. The second part of the study again started with a placebo intervention for 1 week, followed by 6 additional weeks of treatment with the active drug. Nabilone treatment was associated with a significant decrease in agitation, besides important reduction in related caregiver burden. Sedation constituted the most common emerging adverse event, which deserves particular consideration.

Open-label and observational studies

In an open-label pilot study, Walther et al.2222. Walther S, Mahlberg R, Eichmann U, Kunz D. Delta-9-tetrahydrocannabinol for nighttime agitation in severe dementia. Psychopharmacology. 2006;185:524-8. examined the effects of dronabinol for six patients, aged 81.5 years (five with AD and one with VD at advanced stages of dementia) who suffered from behavioral and day-night rhythm disturbances. Patients received 2.5 mg of dronabinol daily for 2 weeks. Nighttime behavior was assessed using actigraphy. With this treatment, agitation and nocturnal motor activity decreased from baseline in all patients. Furthermore, irritability, anxiety and appetite disorders also improved. However, delusions, hallucinations, and apathy did not change during dronabinol therapy. The drug was well tolerated, with no reported adverse events during the study.

Ten consecutive inpatients (mean age 73.2 years) suffering from moderate to severe AD dementia completed a prospective open-label trial, conducted for a 4-week period to examine medical cannabis oil (MCO) for treatment of behavioral disturbances, agitation and aggression.2323. Shelef A, Barak Y, Berger U, Paleacu D, Tadger S, Plopsky I, et al. Safety and efficacy of medical cannabis oil for behavioral and psychological symptoms of dementia: an-open label, add-on, pilot study. J Alzheimers Dis. 2016;51:15-9. The MCO contained THC extract from the Cannabis plant. Chemical analysis of the MCO signalized very low levels of some other phytocannabinoids, e.g., CBD: 0.05%; cannabichromes: 0.05%; cannabinol: 0.17%; tetrahydrocannabivarin: 0.02%; and tetrahydrocannabinolic acid: < 0.01%. The patients received MCO twice a day for 4 weeks, each dose initially containing 2.5 mg of THC. On the 3rd day the dosage was increased two-fold to the equivalent of 5.0 mg of THC b.i.d. ( bis in die , twice a day), and subsequently to 7.5 mg b.i.d. in the absence of side effects or safety concerns. The authors observed a significant decrease in agitation and aggression, but also in aberrant motor behavior, delusions, sleep disorder and nighttime behavior, irritability, and apathy. Additionally, caregiver distress was reduced. Three patients presented clinically relevant side effects: dysphagia (presumably unrelated to the treatment); recurrent falls (which were also present prior to the treatment); and confusion (which improved with reduction of the experimental drug from 5.0 mg to 2.5 mg b.i.d.).

In an observational study, Broers et al.2424. Broers B, Patà Z, Mina A, Wampfler J, de Saussure C, Pautex S. Prescription of a THC/CBD-Based medication to patients with dementia: a pilot study in Geneva. Med Cannabis Cannabinoids. 2019;2:56-9. examined the clinical efficacy and administration feasibility of a THC/CBD-based medication for management of NPS in a nursing home. They carried out a pilot study with 10 women suffering from severe dementia caused by different etiologies (AD, VD, and AD/VD) and presenting with serious behavioral disturbances and some motor rigidity. Patients received oral doses of the THC/CBD preparation, with acceptable tolerability. During the first 2 weeks of the trial, the average daily dosage prescribed to patients was 7.6 mg/13.2 mg of THC/CBD respectively, with titration up to 9.0 mg/18.0 mg over the following 2 months, with stable doses thereafter. The lowest daily dosage was 7.0 mg of THC and 14.0 mg of CBD, and the highest dosage was 13.0 mg and 26.0 mg, respectively. The authors reported benefits in overall behavioral disturbances and motor rigidity. Likewise, the staff members reported benefits in engagement with daily activities and reduction of troublesome behaviors (such as aggression, screaming, tearing clothes) in most patients, who appeared to be calmer, more relaxed, less irritable, and smiling more.

Case reports

Favorable outcomes from the experience with medical cannabinoids were also described in case reports. Passmore1616. Passmore MJ. The cannabinoid receptor agonist nabilone for the treatment of dementia-related agitation. Int J Geriatr Psychiatry. 2008;23:116-7. described the case of a 72 year-old man with severe AD who was transferred from his house to a nursing home because of persistent motor hyperactivity, disinhibition (indiscriminate disrobing and toileting), and verbal physical aggression towards co-residents and staff, and who was unresponsive to routine pharmacological approaches. After treatment with nabilone 0.5 mg b.i.d., he experienced a prompt and sustained reduction in the severity of agitation and aggression, with no emergent adverse events, and was discharged from the nursing home. His NPS remained controlled over the following 3 months.

Another interesting report, published by Amanullah et al.,2626. Amanullah S, MacDougall K, Sweeney N, Coffin J, Cole J. Synthetic cannabinoids in dementia with agitation: case studies and literature review. Clin Neuropsychiatry. 2013;10:142-7. describes the case of a 79-year old man diagnosed with advanced dementia due to AD presenting with increased agitation and caregiver burden, which caused nursing home placement. Upon admission, the patient was undergoing regular pharmacological treatment with donepezil, memantine, and antipsychotics with poor response in relation to the occurrence of NPS. The patient was medicated with haloperidol and quetiapine, but subsequently developed an episode of delirium that resolved after withdrawal of these psychotropics, with persistence of agitation and aggressive behavior. Thereafter, nabilone 0.5 mg was started and titrated to 0.5 mg t.i.d., resulting in long-lasting improvement of agitation and communication, as reported by the staff, who observed that the patient recovered a minimum ability to express himself using short sentences, became acquiescent to verbal command, and displayed emotional reactions, more frequent smiles, and increased eye contact. In the same publication, the authors also reported the case of a 60-year-old man with severe FTD with aggressive behavior and sleep disorders, requiring three-to-four employees for daily hospice care. The patient was on a regular psychotropic regimen with no benefits. Nabilone 0.5 mg b.i.d. was prescribed in association with zopiclone at bedtime, with substantial decrease in aggressiveness, and reduction (to one) of the number of staff members needed to provide daily care. The patient also began to express himself in single words and short sentences. Daily doses of nabilone were increased to 0.5 mg t.i.d. during follow-up due to relapse of aggressive behavior, along with increments in quetiapine up to 300 mg/day. Under this regimen, the patient became progressively less agitated and more cooperative with provision of personal care. The authors concluded that use of nabilone in combination with other medications provoked a sustained reduction of agitation and aggressive behavior.

Defrancesco and Hofer2929. Defrancesco M, Hofer A. Cannabinoid as beneficial replacement therapy for psychotropics to treat neuropsychiatric symptoms in severe Alzheimer’s dementia: a clinical case report. Front Psychiatry. 2020;11:413. published another case report on dronabinol for treatment of NPS in a 79-year-old woman with severe AD. The patient was unresponsive to conventional psychopharmacological intervention, leading to polypharmacy, which in turn caused sedation and other relevant adverse effects. She underwent a progressive worsening of agitation, aggression, anxiety, and disruptive behavior. Dronabinol (magistral prescription of THC) was started at low dose and titrated to THC equivalents of 4.9-6.7 mg/day, with a considerable reduction of symptoms. Her emotional state improved, allowing nursing care without anxious or aggressive behavior, without side effects. The patient maintained the improvement for at least 10 months, until the current report was written, with a low dosage of dronabinol and optimization of other psychotropics at lower doses.

In order to treat a male inpatient with LBD who presented with severe outbursts of aggression with violence against other patients and staff members, Wilhelm et al.2828. Wilhelm R, Ahl B, Anghelescu I-G. Dextrometorphan-paroxetine, but not dronabinol, effective for treatment-resistant aggression and agitation in an elderly patient with lewy body dementia. J Clin Psychopharmacol. 2017;37:745-7. prescribed dronabinol in progressive dosages, starting from 2.5 mg to 15.0 mg per day since conventional drugs had no efficacy. At lower prescriptions, there was no favorable effect, and with increasing doses, the agitation worsened. The treatment was changed to 24.0 mg dextromethorphan and 10.0 mg paroxetine, and after 1 day the patient showed a substantial improvement in aggressive behavior. Unfortunately, the patient killed himself by hanging 6 weeks after referral, but the authors argued that this event was not clearly associated with current medications.

Walther et al.2525. Walther S, Schüpbach B, Seifritz E, Homan P, Strik W. Randomized, controlled crossover trial of dronabinol, 2.5 mg, for agitation in 2 patients with dementia. J Clin Psychopharmacol. 2011;31:256-8. described two treatment approaches with dronabinol for NPS: a 75-year-old man (patient A) and an 81-year-old man (patient B) with moderate AD presenting with circadian rhythm disorders, verbal agitation, and aggression. The patients were prescribed either dronabinol 2.5 mg/day for the first 2 weeks and placebo for the following 2 weeks (patient A), or the other way round (patient B). In the former case, nocturnal motor activity consistently decreased until the 3rd week of treatment, but symptoms returned to the baseline levels at the end of the placebo phase. As for patient B, who received placebo in the first 2 weeks and dronabinol in weeks 3 and 4, nocturnal motor activity was unchanged during the placebo phase and attenuated in the 1st week of dronabinol treatment, relapsing again in the 2nd week of dronabinol. Both patients tolerated the dronabinol treatment well, with no occurrences of severe adverse events.

Sleep disturbances

Case report

Effects of dronabinol on sleep disturbances were also reported for patients A and B in the aforementioned study by Walther et al.2525. Walther S, Schüpbach B, Seifritz E, Homan P, Strik W. Randomized, controlled crossover trial of dronabinol, 2.5 mg, for agitation in 2 patients with dementia. J Clin Psychopharmacol. 2011;31:256-8. Both patients’ circadian rhythms were strengthened and there was a short-lived effect of dronabinol reducing nighttime activity for patient B, i.e., the effect waned after 1 week of treatment. The authors suggested that higher doses of dronabinol might be needed for a sustained improvement of nocturnal behavioral disturbances. The beneficial effect of dronabinol on sleep regulation is presumably related to modulation of the eCB by stimulation of CB1 receptors in the brainstem and subsequent effects on circadian rhythms.3131. Murillo-Rodríguez E. The role of the CB1 receptor in the regulation of sleep. Prog Neuropsychopharmacol Biol Psychiatry. 2008;32:1420-7.

Abnormal sexual behavior

Case report

Zajac et al.2727. Zajac DM, Sikkema SR, Chandrasena R. Nabilone for the treatment of dementia-associated sexual disinhibition. Prim Care Companion CNS Disord. 2015;17: 10.4088/PCC.14l01695.
https://doi.org/10.4088/PCC.14l01695...
report the case of man with mixed vascular and frontotemporal dementia living in a long-term assisted residence who exhibited sexually disinhibited behaviors, such as inappropriate sexual comments, public masturbation, touching the nursing staff, and harassing female residents. Non-pharmacological interventions and several interventions with psychotropic drugs were ineffective.2727. Zajac DM, Sikkema SR, Chandrasena R. Nabilone for the treatment of dementia-associated sexual disinhibition. Prim Care Companion CNS Disord. 2015;17: 10.4088/PCC.14l01695.
https://doi.org/10.4088/PCC.14l01695...
The patient was prescribed nabilone 0.5 to 1.0 mg b.i.d. for 2 weeks and risperidone (0.5 mg orally) when indispensable to control aggressive behavior. The patient achieved a significant improvement in behavioral changes, with complete resolution of sexual disinhibition symptoms. During the treatment, the patient experienced some acute episodes of sedation and lethargy attributed to interaction of nabilone with other sedative drugs, or to a common adverse effect of the medication. When nabilone treatment was eventually interrupted, inappropriate sexual behaviors recurred, but were controlled again with reintroduction of the medication (0.5 mg t.i.d.) and there were continuous gains along the following 3 months of observation.

The main characteristics of the studies selected are presented in Table 1 .

Table 1
Intervention studies of medical cannabinoids for NPS of dementia

The studies were also summarized according to the level of evidence supporting the effects of the various cannabinoid drugs for treatment of NPS ( Table 2 ). Considering all selected publications, only one randomized-controlled trial (RCT) reported the effect size (0.51; a medium effect; Herrmann et al.2121. Herrmann N, Ruthirakuhan M, Gallagher D, Verhoeff NPLG, Kiss A, Black SE, et al. Randomized placebo-controlled trial of nabilone for agitation in Alzheimer’s disease. Am J Geriatr Psychiatry. 2019;27:1161-73. ); and in general, the statistical significance related to outcomes of studies varied widely.

Table 2
NPS and drug efficacy

Discussion

In spite of the enthusiasm of many clinicians towards use of medical cannabinoids for treatment of agitation and other NPS in dementia, there is still very limited evidence from controlled studies to support this prescription.3232. Weier M, Hall W. The use of cannabinoids in treating dementia. Curr Neurol Neurosci Rep. 2017;17:56. , 3333. Abuhasira R, Ron A, Sikorin I, Novack V. Medical cannabis for older patients - treatment protocol and initial results. J Clin Med. 2019;8:1819. These compounds, including magistral formulations of marijuana extracts and pharmaceutical products containing THC and/or CBD, have been tentatively used to treat difficult cases of NPS/BPSD where more established pharmacological and non-pharmacological strategies had failed to deliver benefits.2323. Shelef A, Barak Y, Berger U, Paleacu D, Tadger S, Plopsky I, et al. Safety and efficacy of medical cannabis oil for behavioral and psychological symptoms of dementia: an-open label, add-on, pilot study. J Alzheimers Dis. 2016;51:15-9. , 3434. Hillen JB, Soulsby N, Alderman C, Caughey GE. Safety and effectiveness of cannabinoids for the treatment of neuropsychiatric symptoms in dementia: a systematic review. Ther Adv Drug Saf. 2019;10: 2042098619846993. Controversial results from controlled trials, along with a limited number of successful clinical experiences, have been reported in the format of case reports or small case series.3434. Hillen JB, Soulsby N, Alderman C, Caughey GE. Safety and effectiveness of cannabinoids for the treatment of neuropsychiatric symptoms in dementia: a systematic review. Ther Adv Drug Saf. 2019;10: 2042098619846993. Although clinically relevant, these communications presumably represent a collection of cases where the use of cannabinoid drugs yielded positive responses, amidst a probably larger number of failed attempts – illustrating the burden of publication bias.3434. Hillen JB, Soulsby N, Alderman C, Caughey GE. Safety and effectiveness of cannabinoids for the treatment of neuropsychiatric symptoms in dementia: a systematic review. Ther Adv Drug Saf. 2019;10: 2042098619846993. Nonetheless, the available studies – controlled or not – substantiate the case for further experimentation and emphasize the urgent need for randomized, placebo-controlled trials conducted with larger and better selected patient samples.3232. Weier M, Hall W. The use of cannabinoids in treating dementia. Curr Neurol Neurosci Rep. 2017;17:56. There is also need for better characterization of the behavioral symptoms that may be most responsive to the effects of cannabinoid drugs, in addition to detailed profiling of compounds, doses, and treatment duration.3333. Abuhasira R, Ron A, Sikorin I, Novack V. Medical cannabis for older patients - treatment protocol and initial results. J Clin Med. 2019;8:1819. , 3535. Vacaflor BE, Beauchet O, Jarvis E, Schavietto A, Rej S. Mental health and cognition in older cannabis users: a review. Can Geriatr J. 2020;23:242-9.

In the present study, we analyzed the available publications that were systematically retrieved from the literature according to the criteria established, using MeSH terms clustered into three classificatory domains, i.e., “cognitive impairment/dementia,” “NPS/BPSD,” and “cannabinoid drugs.” The total number of studies reporting original clinical data about use of medical cannabinoids for treatment of NPS was small (n = 15), compared to a 50-fold larger number of publications on this very topic, which obviously include pre-clinical and neurobiological studies, reviews of the literature, and position papers disclosing expert points of view. We further subdivided the eligible papers into three categories that reflect the level of evidence embedded in these contributions, i.e., controlled clinical trials, open-label or observational studies, and case reports. These selected publications comprised five controlled trials, three open-label/observational studies, and seven case reports, which represent the body of publications on this subject for the time being – not much, but valuable. The level of evidence provided by the different studies will be discussed across the topics “efficacy,” “safety/tolerability,” and “limitations.”

Efficacy

Controlled clinical trials (level of evidence 1) with dronabinol (Mahlberg et al.,1818. Mahlberg R, Walther S. Actigraphy in agitated patients with dementia - actigraphy in agitated patients with dementia: monitoring treatment outcomes. Z Gerontol Geriatr. 2007;40:178-84. ) and nabilone2121. Herrmann N, Ruthirakuhan M, Gallagher D, Verhoeff NPLG, Kiss A, Black SE, et al. Randomized placebo-controlled trial of nabilone for agitation in Alzheimer’s disease. Am J Geriatr Psychiatry. 2019;27:1161-73. (Herrmann et al.2121. Herrmann N, Ruthirakuhan M, Gallagher D, Verhoeff NPLG, Kiss A, Black SE, et al. Randomized placebo-controlled trial of nabilone for agitation in Alzheimer’s disease. Am J Geriatr Psychiatry. 2019;27:1161-73. ) demonstrated a consistent improvement in agitation, nighttime behavior, and aggression in patients with severe AD. Open-label studies (level of evidence 2) with dronabinol,2222. Walther S, Mahlberg R, Eichmann U, Kunz D. Delta-9-tetrahydrocannabinol for nighttime agitation in severe dementia. Psychopharmacology. 2006;185:524-8. MCO,2323. Shelef A, Barak Y, Berger U, Paleacu D, Tadger S, Plopsky I, et al. Safety and efficacy of medical cannabis oil for behavioral and psychological symptoms of dementia: an-open label, add-on, pilot study. J Alzheimers Dis. 2016;51:15-9. and THC/CBD-based medicine2424. Broers B, Patà Z, Mina A, Wampfler J, de Saussure C, Pautex S. Prescription of a THC/CBD-Based medication to patients with dementia: a pilot study in Geneva. Med Cannabis Cannabinoids. 2019;2:56-9. also showed efficacy in controlling agitation and other behavioral disturbances in patients with severe AD, VD, and AD/VD. Broers et al.2424. Broers B, Patà Z, Mina A, Wampfler J, de Saussure C, Pautex S. Prescription of a THC/CBD-Based medication to patients with dementia: a pilot study in Geneva. Med Cannabis Cannabinoids. 2019;2:56-9. prescribed natural cannabis extract, with a THC/CBD ratio of 1:2, and this was a particular feature of their study. In contrast to other authors, they ordered higher dosages, with an average of 9.0 mg THC, while most studies recommended lower dosages, from 2.0 mg to 7.0 mg.3636. Aso E, Ferrer I. Cannabinoids for treatment of Alzheimer’s disease: moving toward the clinic. Front Pharmacol. 2014;5:37. Broers et al.2424. Broers B, Patà Z, Mina A, Wampfler J, de Saussure C, Pautex S. Prescription of a THC/CBD-Based medication to patients with dementia: a pilot study in Geneva. Med Cannabis Cannabinoids. 2019;2:56-9. recognized some weaknesses of their study given its observational design.

Likewise, medical cannabis even provoked substantial improvement in delusions and apathy in moderate and severe AD, with subsequent reduction in caregiver distress, according to an open-label study (level of evidence 2).2323. Shelef A, Barak Y, Berger U, Paleacu D, Tadger S, Plopsky I, et al. Safety and efficacy of medical cannabis oil for behavioral and psychological symptoms of dementia: an-open label, add-on, pilot study. J Alzheimers Dis. 2016;51:15-9. Therefore, case reports (level of evidence 3) revealed improvement with dronabinol in distinct neuropsychiatric domains in severe AD, including agitation and nighttime behaviors,2525. Walther S, Schüpbach B, Seifritz E, Homan P, Strik W. Randomized, controlled crossover trial of dronabinol, 2.5 mg, for agitation in 2 patients with dementia. J Clin Psychopharmacol. 2011;31:256-8. , 2929. Defrancesco M, Hofer A. Cannabinoid as beneficial replacement therapy for psychotropics to treat neuropsychiatric symptoms in severe Alzheimer’s dementia: a clinical case report. Front Psychiatry. 2020;11:413. as well as in agitation and aggression with nabilone.1616. Passmore MJ. The cannabinoid receptor agonist nabilone for the treatment of dementia-related agitation. Int J Geriatr Psychiatry. 2008;23:116-7. , 2626. Amanullah S, MacDougall K, Sweeney N, Coffin J, Cole J. Synthetic cannabinoids in dementia with agitation: case studies and literature review. Clin Neuropsychiatry. 2013;10:142-7.

Notably, in patients with severe FTD, nabilone was effective for control of agitation and aggression,2626. Amanullah S, MacDougall K, Sweeney N, Coffin J, Cole J. Synthetic cannabinoids in dementia with agitation: case studies and literature review. Clin Neuropsychiatry. 2013;10:142-7. with reduced disinhibition and sexual disorders.2727. Zajac DM, Sikkema SR, Chandrasena R. Nabilone for the treatment of dementia-associated sexual disinhibition. Prim Care Companion CNS Disord. 2015;17: 10.4088/PCC.14l01695.
https://doi.org/10.4088/PCC.14l01695...
Favorable results were also observed in patients with FTD suffering from agitation and anxiety who were given CBD (medical marijuana).3030. Gopalakrishna G, Srivathsal Y, Kaur G. Cannabinoids in the management of frontotemporal dementia: a case series. Neurodegener Dis Manag. 2021;11:61-4.

Conversely, in two controlled trials (level of evidence 1), enrolling patients with severe AD, VD, and AD/VD, who were suffering from agitation and other NPS, the authors found no significant differences between dronabinol and placebo.1919. van den Elsen GAH, Ahmed AIA, Verkes R-J, Feuth T, van der Marck MA, Olde Rikkert MGM. Tetrahydrocannabinol in behavioral disturbances in dementia: a crossover randomized controlled trial. Am J Geriatr Psychiatry. 2015;23:1214-24a. , 2020. van den Elsen GAH, Ahmed AIA, Verkes R-J, Kramers C, Feuth T, Rosenberg PB, et al. Tetrahydrocannabinol for neuropsychiatric symptoms in dementia: a randomized controlled trial. Neurology. 2015;84:2338-46b. Moreover, one of the patients (“B”) with severe AD2525. Walther S, Schüpbach B, Seifritz E, Homan P, Strik W. Randomized, controlled crossover trial of dronabinol, 2.5 mg, for agitation in 2 patients with dementia. J Clin Psychopharmacol. 2011;31:256-8. who received dronabinol, achieved an improvement in nighttime agitation for only a very short period. Additionally, two clinical trials with this drug showed no benefits for NPS. Along the same lines, a patient with LBD and resistant aggressive behavior had no response to dronabinol, and actually got even worse with the treatment.2828. Wilhelm R, Ahl B, Anghelescu I-G. Dextrometorphan-paroxetine, but not dronabinol, effective for treatment-resistant aggression and agitation in an elderly patient with lewy body dementia. J Clin Psychopharmacol. 2017;37:745-7.

Woodward et al.3737. Woodward MR, Harper DG, Stolyar A, Forester BP, Ellison JM. Dronabinol for the treatment of agitation and aggressive behavior in acutely hospitalized severely demented patients with noncognitive behavioral symptoms. Am J Geriatr Psychiatry. 2014;22:415-9. conducted a retrospective analysis based on medical records in order to detect the efficacy and tolerability of dronabinol for treatment of inpatients with severe dementia from different etiologies (AD, VD, AD/VD, and FTD). Clinically relevant behavioral disturbances, e.g., agitation, aggression, sleep disorders, and anorexia were the main focus of analyses. The authors concluded that dronabinol may be an adjunctive strategy for treatment of NPS in dementia. It is plausible to draw attention to favorable effects of medical cannabinoids on regulation of circadian rhythm and sleep architecture, as well as in controlling nocturnal behaviors of patients with advanced stages of AD.3838. Gates PJ, Albertella L, Copeland J. The effects of cannabinoid administration on sleep: a systematic review of human studies. Sleep Med Rev. 2014;18:477-87. , 3939. Suryadevara U, Bruijnzeel DM, Nuthi M, Jagnarine DA, Tandon R, Bruijnzeel AW. Pros and cons of medical cannabis use by people with chronic brain disorders. Curr Neuropharmacol. 2017;15:800-14.

According to Murillo-Rodriguez,3131. Murillo-Rodríguez E. The role of the CB1 receptor in the regulation of sleep. Prog Neuropsychopharmacol Biol Psychiatry. 2008;32:1420-7. the effects of dronabinol on circadian rhythm can be related to the eCB, which modulates sleep related-properties via CB1 receptors in the brainstem. Furthermore, experimental investigations represent a basis for new clinical trials aimed at expanding knowledge about the potential benefits of medical cannabinoids. Notably, THC decreases agonistic acts in multiple mammalian species (mice, rats, and squirrel monkeys) undergoing intruder confrontation.4040. Miczek K. delta9-tetrahydrocannabinol: antiaggressive effects in mice, rats, and squirrel monkeys. Science. 1978;199:1459-61. , 4141. Paronis CA, Nikas SP, Shukla VG, Makriyannis A. Δ(9)-Tetrahydrocannabinol acts as a partial agonist/antagonist in mice. Behav Pharmacol. 2012;23:802-5. Mutant mice lacking CB1 exhibit more aggressive or defensive behavior, such as avoidance, freezing, and risk-assessment actions, suggesting CB1 plays a role in buffering against social stress.4242. Rodriguez-Arias M, Navarrete F, Daza-Losada M, Navarro D, Aguilar MA, Berbel P, et al. CB1 cannabinoid receptor-mediated aggressive behavior. Neuropharmacology. 2013;75:172-80. , 4343. Litvin Y, Phan A, Hill MN, Pfaff DW, McEwen BS. CB1 receptor signaling regulates social anxiety and memory. Genes Brain Behav. 2013;12:479-89. The statistical significance varied widely between studies in our analysis, both according to the results obtained by different assessments, as well as within the same scale when measuring selected psychopathological domains.

Concerning NPS related to different entities in other populations, medical cannabinoids have also shown benefits. For instance, according to a review on CBD treatment of non-motor symptoms of Parkinson’s disease, patients who received this compound exhibited significant improvement in psychosis and sleep behavior disorder, as well as better performance in daily activities, and became more able to deal with the stigma related to the disease.4444. Crippa JAS, Hallak JEC, Zuardi AW, Guimarães FS, Tumas V, Santos RG. Is the cannabidiol the ideal drug to treat non-motor Parkinson’s disease symptoms? Eur Arch Psychiatry Clin Neurosci. 2019;269:121-33. Furthermore, patients with severe treatment-resistant epilepsy for whom CBD was prescribed in addition to antiepileptic drugs, exhibited an improvement in behavioral disorders and in quality of life and also achieved a reduced frequency of seizures.4545. Silvestro S, Mammana S, Cavalli E, Bramanti P, Mazzon E. Use of cannabidiol in the treatment of epilepsy: efficacy and security in clinical trials. Molecules. 2019;24:1459. An important methodological weakness of studies carried out in different populations, including our investigation, concerns the small sample size and short-term interventions. Additionally, a considerable number of clinical studies on medical cannabinoids for treatment of NPS in dementia and other clinical entities, have failed or have not reached a decisive agreement on their effectiveness.1919. van den Elsen GAH, Ahmed AIA, Verkes R-J, Feuth T, van der Marck MA, Olde Rikkert MGM. Tetrahydrocannabinol in behavioral disturbances in dementia: a crossover randomized controlled trial. Am J Geriatr Psychiatry. 2015;23:1214-24a. , 2020. van den Elsen GAH, Ahmed AIA, Verkes R-J, Kramers C, Feuth T, Rosenberg PB, et al. Tetrahydrocannabinol for neuropsychiatric symptoms in dementia: a randomized controlled trial. Neurology. 2015;84:2338-46b. , 2828. Wilhelm R, Ahl B, Anghelescu I-G. Dextrometorphan-paroxetine, but not dronabinol, effective for treatment-resistant aggression and agitation in an elderly patient with lewy body dementia. J Clin Psychopharmacol. 2017;37:745-7. , 3333. Abuhasira R, Ron A, Sikorin I, Novack V. Medical cannabis for older patients - treatment protocol and initial results. J Clin Med. 2019;8:1819. , 3434. Hillen JB, Soulsby N, Alderman C, Caughey GE. Safety and effectiveness of cannabinoids for the treatment of neuropsychiatric symptoms in dementia: a systematic review. Ther Adv Drug Saf. 2019;10: 2042098619846993. , 4646. Bahji A, Meyyappan AC, Hawken E. Cannabinoids for the neuropsychiatric symptoms of edmentia: a systematic review and meta-analysis. Can J Psychiatry. 2020;65:365-76.

Safety and tolerability

Regarding safety and tolerability, participants in general presented mild side effects with dronabinol, nabilone, and other cannabinoid compounds. Dronabinol and nabilone can cause transient sedation, somnolence, and mild euphoria, possibly related to interaction with basic-routine drugs. One patient presented lethargy and delirium during nabilone use in the context of polypharmacy, which at least in part could explain this event. Another one had a generalized seizure during dronabinol use, but it was not clear whether the event was caused by the drug or by progression of the AD pathology itself. In this context, Zajac et al.2727. Zajac DM, Sikkema SR, Chandrasena R. Nabilone for the treatment of dementia-associated sexual disinhibition. Prim Care Companion CNS Disord. 2015;17: 10.4088/PCC.14l01695.
https://doi.org/10.4088/PCC.14l01695...
emphasized the risk of sedation, lethargy, and delirium in patients with dementia when prescribed medical cannabinoids. Woodward et al.3737. Woodward MR, Harper DG, Stolyar A, Forester BP, Ellison JM. Dronabinol for the treatment of agitation and aggressive behavior in acutely hospitalized severely demented patients with noncognitive behavioral symptoms. Am J Geriatr Psychiatry. 2014;22:415-9. observed that such events either occurred amidst pharmacological interactions with other drugs in use, or could be related to general medical comorbidities. Defrancesco and Hofer2929. Defrancesco M, Hofer A. Cannabinoid as beneficial replacement therapy for psychotropics to treat neuropsychiatric symptoms in severe Alzheimer’s dementia: a clinical case report. Front Psychiatry. 2020;11:413. draw attention to the fact that the hepatic metabolism of THC by cytochrome P450 isoenzymes can cause a considerable risk of drug-drug interactions. Although in general cannabinoids are well tolerated, it is mandatory to monitor potential side effects given the marked vulnerability of patients with dementia.

The safety and tolerability of medical cannabinoids has also been the subject of a recent comprehensive review of randomized controlled trials covering populations other than those with dementia from different etiologies.4747. Velayudhan L, McGoohan K, Bhattacharyya S. Safety and tolerability of natural and synthetic cannabinoids in adults aged over 50 years: a systematic review and meta-analysis. PLoS Med. 2021;18:e1003524. The review included patients with Parkinson’s disease, multiple sclerosis, Huntington disease, advanced cancer with pain, rheumatoid arthritis, and diabetes, among others. The authors reported no significant increase in serious events and recognized medical cannabinoids in general as a safe and relatively well-tolerated treatment in older adults.4747. Velayudhan L, McGoohan K, Bhattacharyya S. Safety and tolerability of natural and synthetic cannabinoids in adults aged over 50 years: a systematic review and meta-analysis. PLoS Med. 2021;18:e1003524. Likewise, according to a review conducted by Crippa et al.4444. Crippa JAS, Hallak JEC, Zuardi AW, Guimarães FS, Tumas V, Santos RG. Is the cannabidiol the ideal drug to treat non-motor Parkinson’s disease symptoms? Eur Arch Psychiatry Clin Neurosci. 2019;269:121-33. in patients with Parkinson’s disease, CBD was well tolerated when prescribed for treatment of non-motor symptoms.

Methodological aspects and limitations

RCTs with adequate sample sizes, reputed as the best approach to validate new therapeutic interventions, are still lacking in this field of knowledge. To date, only six controlled studies have been conducted to test the efficacy and safety of medical cannabinoid drugs for treatment of NPS/BPSD, jeopardizing construction of a solid evidence-based background (level of evidence 1). In addition, in many such studies, the patient samples were small, with fifty participants or less. Three studies had an open-label design (level 2), also with small sample sizes ranging from six to 10 subjects. By definition, case reports (level 3) enrolled even smaller numbers of patients. Heterogeneity of samples is another important issue, since most of the aforementioned studies comprised patients with dementia of different etiologies (e.g., AD, VD, AD/VD, FTD, and DLB), with distinct profiles of neurobehavioral disturbances. Such variability within groups may have impacted on outcomes and responses to treatment, restricting the comparability of studies and extrapolation of findings. Such methodological characteristics have also been pointed out by previous review studies on NPS in older populations.3434. Hillen JB, Soulsby N, Alderman C, Caughey GE. Safety and effectiveness of cannabinoids for the treatment of neuropsychiatric symptoms in dementia: a systematic review. Ther Adv Drug Saf. 2019;10: 2042098619846993. , 4646. Bahji A, Meyyappan AC, Hawken E. Cannabinoids for the neuropsychiatric symptoms of edmentia: a systematic review and meta-analysis. Can J Psychiatry. 2020;65:365-76. , 4747. Velayudhan L, McGoohan K, Bhattacharyya S. Safety and tolerability of natural and synthetic cannabinoids in adults aged over 50 years: a systematic review and meta-analysis. PLoS Med. 2021;18:e1003524.

It is also worth mentioning that controlled trials and open-label studies were characterized by different periods of intervention, extending from 2 to 14 weeks. Case studies also varied widely, from 7 to 78 days. Likewise, the assessment of outcomes differs from study to study, most lacking more comprehensive and longitudinal appraisals. Most patients were classified as having advanced dementia, and a smaller group presented moderate dementia. These data are clinically relevant because NPS, especially agitation, nighttime behavior, wandering, sleep disorders, eating disorders, sexual disinhibition, and apathy tend to worsen in the more advanced stages of dementia, increasing caregiver burden.3333. Abuhasira R, Ron A, Sikorin I, Novack V. Medical cannabis for older patients - treatment protocol and initial results. J Clin Med. 2019;8:1819. , 4848. Shim SH, Kang HS, Kim JH, Kim DK. Factors associated with caregiver burden in dementia: 1-year follow-up study. Psychiatry Investig. 2016;13(1):43-49. , 4949. Magierski R, Sobow T, Schwertner E, Religa D. Pharmacotherapy of behavioral and psychological symptoms of dementia: state of the art and future progress. Front Pharmacol. 2020;11:1168. Another aspect to be considered regards variation in the dosage regimens of different compounds. Some studies adopted fixed doses of the cannabinoid drugs, while others used variable prescriptions throughout the investigation. This fact suggests that the therapeutic window for this class of drugs is yet to be defined. Notably, the dosing schedule for medical cannabinoids aims to determine the best efficacy of the compound and, at the same time, maintain an acceptable level of possible adverse events.5050. Ahmed AIA, van den Elsen GAH, Colbers A, Kramers C, Burger DM, van der Marck MA, et al. Safety, pharmacodynamics, and pharmacokinetics of multiple oral doses of delta-9-tetrahydrocannabinol in older persons with dementia. Psychopharmacology. 2015;232:2587-95.

Several aspects were related to the decision to restrict our study to a systematic review without a meta-analysis of primary outcomes. Factors that precluded the meta-analytical approach were: incomplete data reported in the original manuscripts, heterogeneous methodological designs, different drug dosages, different delivery routes adopted for therapy, variation in the percentage of pharmacological compounds of medical cannabinoids, and different periods of treatment. Another limitation of our study is that we did not register the review and a protocol was not prepared in advance. Further trials incorporating methodological designs based on larger and longer-term investigations are strongly advocated to determine the real-world safety and effectiveness of medical cannabinoids. Studies should establish an appropriate dosage-escalation regimen to maximize efficacy while at the same time keeping adverse events acceptable, in particular for fragile patients with associated comorbidities. Another critical point concerns explanation of neurobiological mechanisms and the pharmacological actions of cannabinoids within the brain that are involved in behavior regulation.

Conclusion

Cannabinoid-based drugs are a promising psychopharmacological strategy to treat patients with NPS related to moderated or advanced dementia. In general, studies with medical cannabinoids have shown favorable results, although some do not provide convincing efficacy. Basically, compounds are well tolerated, but given the vulnerability of patients with dementia they require appropriate monitoring by the clinician. Development of controlled trials with longitudinal design and larger samples is needed to determine their long-term efficacy, adverse events, risks of interactions with other drugs, and respective dosage ranges, as well as the most suitable therapy period. Within this approach, determining the respective THC/CBD concentrations for specific drugs to treat different psychopathological domains in dementia still remains an additional concern.

References

  • 1
    Steinberg M, Shao H, Zandi P, Lyketsos CG, Welsh-Bohmer KA, Norton MC, et al. Point and 5-year period prevalence of neuropsychiatric symptoms in dementia: the Cache County Study. Int J Geriatr Psychiatry. 2008;23:170-77.
  • 2
    Lyketsos CG. Neuropsychiatric symptoms in dementia: overview and measurement challenges. J Prev Alzheimers Dis. 2015;2:155-6.
  • 3
    Radue R, Walaszek A, Asthana S. Neuropsychiatric symptoms in dementia. Handb Clin Neurol. 2019;167:437-54.
  • 4
    Jost BC, Grossberg GT. The evolution of psychiatric symptoms in Alzheimer’s disease: a natural history study. J Am Geriatr Society. 1996;44:1078-81.
  • 5
    Mega MS, Cummings JL, Fiorello T, Gornbein J. The spectrum of behavioral changes in Alzheimer’s. Neurology. 1996;46:130-5.
  • 6
    Stella F, Radanovic M, Balthazar MLF, Canineu PR, de Souza LC, Forlenza OV. Neuropsychiatric symptoms in the prodromal stages of dementia. Curr Opin Psychiatry.2014;27:230-5.
  • 7
    Boccardi V, Conestabile Della Staffa M, Baroni M, Ercolani S, Croce MF, Ruggiero C, et al. Prevalence and correlates of behavioral disorders in old age subjects with cognitive impairment: results from the ReGAl project. J Alzheimers Dis. 2017;60:1275-83.
  • 8
    Maust DT, Kim HM, Seyfried LS, Chiang C, Kavanagh J, Schneider LS, et al. Antipsychotics, other psychotropics, and the risk of death in patients with dementia: number needed to harm. JAMA Psychiatry. 2015;72:438-45.
  • 9
    Kales HC, Lyketsos CG, Miller EM, Ballard C. Management of behavioral and psychological symptoms in people with Alzheimer’s disease: an international Delphi consensus. Int Psychogeriatr. 2019;31:83-90.
  • 10
    Mueller C, John C, Perera G, Aarsland D, Ballard C, Stewart R. Antipsychotic use in dementia: the relationship between neuropsychiatric symptom profiles and adverse outcomes. Eur J Epidemiol. 2021;36:89-101.
  • 11
    Brodaty H, Arasaratnam C. Meta-analysis of nonpharmacological interventions for neuropsychiatric symptoms of dementia. Am J Psychiatry. 2012;169:946-53.
  • 12
    Fernández-Ruiz J, Pazos MR, García-Arencibia M, Sagredo O, Ramos JA. Role of CB2 receptors in neuroprotective effects of cannabinoids. Mol Cell Endocrinol. 2008;286:S91-6.
  • 13
    Busquets-Garcia A, Bains J, Marsicano G. CB1 Receptor signaling in the brain: extracting specificity from ubiquity. Neuropsychopharmacology. 2018;43:4-20.
  • 14
    Navarro G, Morales P, Rodríguez-Cueto C, Fernández-Ruiz J, Jagerovic N, Franco R. Targeting cannabinoid CB2 receptors in the central nervous system. medicinal chemistry approaches with focus on neurodegenerative disorders. Front Neurosci. 2016;10:406.
  • 15
    Burggren AC, Shirazi A, Ginder N, London ED. Cannabis effects on brain structure, function, and cognition: considerations for medical uses of cannabis and its derivatives. Am J Drug Alcohol Abuse. 2019;45:563-79.
  • 16
    Passmore MJ. The cannabinoid receptor agonist nabilone for the treatment of dementia-related agitation. Int J Geriatr Psychiatry. 2008;23:116-7.
  • 17
    Volicer L, Stelly M, Morris J, McLaughlin J, Volicer BJ. Effects of dronabinol on anorexia and disturbed behavior in patients with Alzheimer’s disease. Int J Geriatr Psychiatry. 1997;12:913-9.
  • 18
    Mahlberg R, Walther S. Actigraphy in agitated patients with dementia - actigraphy in agitated patients with dementia: monitoring treatment outcomes. Z Gerontol Geriatr. 2007;40:178-84.
  • 19
    van den Elsen GAH, Ahmed AIA, Verkes R-J, Feuth T, van der Marck MA, Olde Rikkert MGM. Tetrahydrocannabinol in behavioral disturbances in dementia: a crossover randomized controlled trial. Am J Geriatr Psychiatry. 2015;23:1214-24a.
  • 20
    van den Elsen GAH, Ahmed AIA, Verkes R-J, Kramers C, Feuth T, Rosenberg PB, et al. Tetrahydrocannabinol for neuropsychiatric symptoms in dementia: a randomized controlled trial. Neurology. 2015;84:2338-46b.
  • 21
    Herrmann N, Ruthirakuhan M, Gallagher D, Verhoeff NPLG, Kiss A, Black SE, et al. Randomized placebo-controlled trial of nabilone for agitation in Alzheimer’s disease. Am J Geriatr Psychiatry. 2019;27:1161-73.
  • 22
    Walther S, Mahlberg R, Eichmann U, Kunz D. Delta-9-tetrahydrocannabinol for nighttime agitation in severe dementia. Psychopharmacology. 2006;185:524-8.
  • 23
    Shelef A, Barak Y, Berger U, Paleacu D, Tadger S, Plopsky I, et al. Safety and efficacy of medical cannabis oil for behavioral and psychological symptoms of dementia: an-open label, add-on, pilot study. J Alzheimers Dis. 2016;51:15-9.
  • 24
    Broers B, Patà Z, Mina A, Wampfler J, de Saussure C, Pautex S. Prescription of a THC/CBD-Based medication to patients with dementia: a pilot study in Geneva. Med Cannabis Cannabinoids. 2019;2:56-9.
  • 25
    Walther S, Schüpbach B, Seifritz E, Homan P, Strik W. Randomized, controlled crossover trial of dronabinol, 2.5 mg, for agitation in 2 patients with dementia. J Clin Psychopharmacol. 2011;31:256-8.
  • 26
    Amanullah S, MacDougall K, Sweeney N, Coffin J, Cole J. Synthetic cannabinoids in dementia with agitation: case studies and literature review. Clin Neuropsychiatry. 2013;10:142-7.
  • 27
    Zajac DM, Sikkema SR, Chandrasena R. Nabilone for the treatment of dementia-associated sexual disinhibition. Prim Care Companion CNS Disord. 2015;17: 10.4088/PCC.14l01695.
    » https://doi.org/10.4088/PCC.14l01695
  • 28
    Wilhelm R, Ahl B, Anghelescu I-G. Dextrometorphan-paroxetine, but not dronabinol, effective for treatment-resistant aggression and agitation in an elderly patient with lewy body dementia. J Clin Psychopharmacol. 2017;37:745-7.
  • 29
    Defrancesco M, Hofer A. Cannabinoid as beneficial replacement therapy for psychotropics to treat neuropsychiatric symptoms in severe Alzheimer’s dementia: a clinical case report. Front Psychiatry. 2020;11:413.
  • 30
    Gopalakrishna G, Srivathsal Y, Kaur G. Cannabinoids in the management of frontotemporal dementia: a case series. Neurodegener Dis Manag. 2021;11:61-4.
  • 31
    Murillo-Rodríguez E. The role of the CB1 receptor in the regulation of sleep. Prog Neuropsychopharmacol Biol Psychiatry. 2008;32:1420-7.
  • 32
    Weier M, Hall W. The use of cannabinoids in treating dementia. Curr Neurol Neurosci Rep. 2017;17:56.
  • 33
    Abuhasira R, Ron A, Sikorin I, Novack V. Medical cannabis for older patients - treatment protocol and initial results. J Clin Med. 2019;8:1819.
  • 34
    Hillen JB, Soulsby N, Alderman C, Caughey GE. Safety and effectiveness of cannabinoids for the treatment of neuropsychiatric symptoms in dementia: a systematic review. Ther Adv Drug Saf. 2019;10: 2042098619846993.
  • 35
    Vacaflor BE, Beauchet O, Jarvis E, Schavietto A, Rej S. Mental health and cognition in older cannabis users: a review. Can Geriatr J. 2020;23:242-9.
  • 36
    Aso E, Ferrer I. Cannabinoids for treatment of Alzheimer’s disease: moving toward the clinic. Front Pharmacol. 2014;5:37.
  • 37
    Woodward MR, Harper DG, Stolyar A, Forester BP, Ellison JM. Dronabinol for the treatment of agitation and aggressive behavior in acutely hospitalized severely demented patients with noncognitive behavioral symptoms. Am J Geriatr Psychiatry. 2014;22:415-9.
  • 38
    Gates PJ, Albertella L, Copeland J. The effects of cannabinoid administration on sleep: a systematic review of human studies. Sleep Med Rev. 2014;18:477-87.
  • 39
    Suryadevara U, Bruijnzeel DM, Nuthi M, Jagnarine DA, Tandon R, Bruijnzeel AW. Pros and cons of medical cannabis use by people with chronic brain disorders. Curr Neuropharmacol. 2017;15:800-14.
  • 40
    Miczek K. delta9-tetrahydrocannabinol: antiaggressive effects in mice, rats, and squirrel monkeys. Science. 1978;199:1459-61.
  • 41
    Paronis CA, Nikas SP, Shukla VG, Makriyannis A. Δ(9)-Tetrahydrocannabinol acts as a partial agonist/antagonist in mice. Behav Pharmacol. 2012;23:802-5.
  • 42
    Rodriguez-Arias M, Navarrete F, Daza-Losada M, Navarro D, Aguilar MA, Berbel P, et al. CB1 cannabinoid receptor-mediated aggressive behavior. Neuropharmacology. 2013;75:172-80.
  • 43
    Litvin Y, Phan A, Hill MN, Pfaff DW, McEwen BS. CB1 receptor signaling regulates social anxiety and memory. Genes Brain Behav. 2013;12:479-89.
  • 44
    Crippa JAS, Hallak JEC, Zuardi AW, Guimarães FS, Tumas V, Santos RG. Is the cannabidiol the ideal drug to treat non-motor Parkinson’s disease symptoms? Eur Arch Psychiatry Clin Neurosci. 2019;269:121-33.
  • 45
    Silvestro S, Mammana S, Cavalli E, Bramanti P, Mazzon E. Use of cannabidiol in the treatment of epilepsy: efficacy and security in clinical trials. Molecules. 2019;24:1459.
  • 46
    Bahji A, Meyyappan AC, Hawken E. Cannabinoids for the neuropsychiatric symptoms of edmentia: a systematic review and meta-analysis. Can J Psychiatry. 2020;65:365-76.
  • 47
    Velayudhan L, McGoohan K, Bhattacharyya S. Safety and tolerability of natural and synthetic cannabinoids in adults aged over 50 years: a systematic review and meta-analysis. PLoS Med. 2021;18:e1003524.
  • 48
    Shim SH, Kang HS, Kim JH, Kim DK. Factors associated with caregiver burden in dementia: 1-year follow-up study. Psychiatry Investig. 2016;13(1):43-49.
  • 49
    Magierski R, Sobow T, Schwertner E, Religa D. Pharmacotherapy of behavioral and psychological symptoms of dementia: state of the art and future progress. Front Pharmacol. 2020;11:1168.
  • 50
    Ahmed AIA, van den Elsen GAH, Colbers A, Kramers C, Burger DM, van der Marck MA, et al. Safety, pharmacodynamics, and pharmacokinetics of multiple oral doses of delta-9-tetrahydrocannabinol in older persons with dementia. Psychopharmacology. 2015;232:2587-95.

Publication Dates

  • Publication in this collection
    17 Dec 2021
  • Date of issue
    Oct-Dec 2021

History

  • Received
    16 Apr 2021
  • Accepted
    14 June 2021
Associação de Psiquiatria do Rio Grande do Sul Av. Ipiranga, 5311/202, 90610-001 Porto Alegre RS/ Brasil, Tel./Fax: (55 51) 3024 4846 - Porto Alegre - RS - Brazil
E-mail: trends@aprs.org.br