ArticlePDF Available

Parkinson’s Disease and Dementia with Lewy Bodies, Patients Under Treatment with Standardized Extracts Enriched in Cannabidiol and Cannabigerol: Descriptive Observations in Preparation for a Phase 2a Clinical Trial

Authors:

Abstract and Figures

Background: Cannabis-based formulations are now widely used by patients with neurological and psychiatric problems but no studies have been published on the clinical utility of CBD/CBG enriched extracts for parkinsonism's symptoms. Objectives: To describe preliminary clinical data collection of PD and DLB patients under CBD/CBG medical prescription. Methods: Review of electronic records of 14 PD and 5 DLB patients. Four extracts were available 1) CBD broad spectrum (100 mg/ mL) 2) CBD/CBG broad spectrum (100 mg/mL 2:1) (3) CBD/CBG (2:1) + THC0.3% full spectrum (100mg/mL) 4) CBD+THC0.3% full spectrum (100 mg/mL). All the patients received authorization from ANVISA (Brazil) to import the formulations for medical use. Outcomes of each unmet need (UMN) were tabulated and graded. Results: Demographics: PD N = 14 (10 male). DLB: N = 5 (3 male). Mean age: PD: 76.2 yrs. (46-94). DLB: 82.2 yrs. (83-92). Disease duration: PD (6.57 yrs.), DLB (4.2 yrs.); PD H/Y stage (3); PD levodopa dose: 490 mg (150-900). Mean daily doses: PD CBD: 65.17 mg (8.33-125 mg), CBG: 22,50 mg (4.16-50 mg), THC: 2,32 mg (0,75-4,5 mg). DLB CBD: 52 mg (5-100 mg). CBG: 8,75 mg (2,5-15 mg), THC: 0,225 mg. Positive results were seen for RBD, insomnia, anxiety, and pain. All pain responders were on CBG and/or THC formulations. Hallucinations were also attenuated in both patient groups. Safety and tolerability were favorable in this small sample. Conclusions: Future clinical trials in Parkinson's disease and DLB with cannabinoids should focus on their potential benefit for associated anxiety, and pain. The potential anti-psychotic effects of CBD and CBD/CBG should also be further evaluated in a phase 2a clinical trial. Abstract Citation: Flávio Henrique de Rezende Costa., et al. "Parkinson's Disease and Dementia with Lewy Bodies, Patients Under Treatment with Standardized
Content may be subject to copyright.
Cronicon
OPEN ACCESS EC NEUROLOGYEC NEUROLOGY
Research Article
Parkinson’s Disease and Dementia with Lewy Bodies, Patients Under
Treatment with Standardized Extracts Enriched in Cannabidiol and
Cannabigerol: Descriptive Observations in Preparation for
a Phase 2a Clinical Trial
Flávio Henrique de Rezende Costa1,2*, Simone Pellegrino1, Mariana Spitz3, Eduardo Rydz1, Gabriel de Castro
Micheli1, Elio Tanaka4, Brian Michael Ebner5, Jaron Gladstone5 and Andrew J Lees6
1Health Meds Laboratories, Rio de Janeiro, Brazil
2Multidisciplinary Clinic, Botagofo, Rio de Janeiro, Brazil
3Estate University of Rio de Janeiro, Brazil
4TNK, Curitiba- Brazil
5CBCeuticals, Coral Springs, Flórida, USA
6Queen Square Brain Bank for Neurological Disorders, University College London Queen Square Institute of Neurology, London, UK
*Corresponding Author: Flávio Henrique de Rezende Costa, Health Meds Laboratories, Rio de Janeiro and Multidisciplinary Clinic,
Botagofo, Rio de Janeiro, Brazil.
Received: December 29, 2021; Published: April 26, 2022
Background: Cannabis-based formulations are now widely used by patients with neurological and psychiatric problems but no stud-
ies have been published on the clinical utility of CBD/CBG enriched extracts for parkinsonism’s symptoms.
Objectives: To describe preliminary clinical data collection of PD and DLB patients under CBD/CBG medical prescription.
Methods: Review of electronic records of 14 PD and 5 DLB patients. Four extracts were available 1) CBD broad spectrum (100 mg/
mL) 2) CBD/CBG broad spectrum (100 mg/mL 2:1) (3) CBD/CBG (2:1) + THC0.3% full spectrum (100mg/mL) 4) CBD+THC0.3%
full spectrum (100 mg/mL). All the patients received authorization from ANVISA (Brazil) to import the formulations for medical use.
Outcomes of each unmet need (UMN) were tabulated and graded.
Results: Demographics: PD N = 14 (10 male). DLB: N = 5 (3 male). Mean age: PD: 76.2 yrs. (46 - 94). DLB: 82.2 yrs. (83 - 92). Disease
duration: PD (6.57 yrs.), DLB (4.2 yrs.); PD H/Y stage (3); PD levodopa dose: 490 mg (150 - 900). Mean daily doses: PD CBD: 65.17
mg (8.33 – 125 mg), CBG: 22,50 mg (4.16 - 50 mg), THC: 2,32 mg (0,75 - 4,5 mg). DLB CBD: 52 mg (5 - 100 mg). CBG: 8,75 mg (2,5 - 15
mg), THC: 0,225 mg. Positive results were seen for RBD, insomnia, anxiety, and pain. All pain responders were on CBG and/or THC
formulations. Hallucinations were also attenuated in both patient groups. Safety and tolerability were favorable in this small sample.
Conclusions: -
sociated anxiety, and pain. The potential anti-psychotic effects of CBD and CBD/CBG should also be further evaluated in a phase 2a
clinical trial.
Keywords: Cannabidiol; Cannabigerol; Parkinson’s Disease; Dementia With Lewy Bodies; Medical Cannabis
Abstract
Citation: Flávio Henrique de Rezende Costa., et al. “Parkinson’s Disease and Dementia with Lewy Bodies, Patients Under Treatment with
Standardized Extracts Enriched in Cannabidiol and Cannabigerol: Descriptive Observations in Preparation for a Phase 2a Clinical Trial”.
EC Neurology 14.5 (2022): 01-15.
DOI: 10.31080/ecne.2022.14.01023
Citation: Flávio Henrique de Rezende Costa., et al. “Parkinson’s Disease and Dementia with Lewy Bodies, Patients Under Treatment with
Standardized Extracts Enriched in Cannabidiol and Cannabigerol: Descriptive Observations in Preparation for a Phase 2a Clinical Trial”.
EC Neurology 14.5 (2022): 01-15.
Abbreviations
ANVISA: National Agency of Health Vigilance (Brazil); ASTM: Accelerated Aging of Sterile Barrier Systems for Medical Devices; CBD: Can-
nabidiol; CBG: Cannabigerol; CPGF: Cannabidiol Pharmaceutical Grade Formulations; DBS: Deep Brain Stimulation; DLB: Dementia with
Lewy Bodies;
GMP: Good Manufactured Practice; ICB: Impulsive, Compulsive Behaviors; MS-HPLC: Mass Spectroscopy – High-Performance Liquid Chro-
matography; MCT oil: Medium Chain Triglycerides Oil;

Introduction
In a A Manual of Diseases of the Nervous System published in 1888 the English neurologist William Gowers described the therapeutic
role of “Indian hemp” (Cannabis indica) sometimes combined with opium- as an effective treatment for tremor in Parkinson’s disease [1].
cannabis had been published [2]. During
the early 20th century, Merck Laboratories (Germany), Burroughs-Wellcome (England), Bristol-Meyers Squibb (USA), Parke-Davis (USA),
and Eli Lilly (USA) had in their portfolios of medications, extracts or tinctures of cannabis [3]. In 1937, the marijuana tax act – contrary
to the American Medical Association’s recommendation- led to the marked decline of medical use of cannabis and in 1941, cannabis was

cannabis resumed in the early ‘60s, with the discovery and synthesis of the principal psychoactive
constituent of Cannabis 
   

Cloning CB1 [6] and CB2 receptors [7] in the early 1990s opened the door to current knowledge about the endocannabinoid system
– or the endocannabinoidome as it is sometimes referred to - an intricate network of biochemically related receptors, enzymes, and me-
diators. The endocannabinoid system’s signaling involves regulating cells, tissues, and organs; organism homeostasis after insults; brain
development and release of neurotransmitters and cytokines related to synaptic plasticity [8].
So far medicinal research has concentrated on CBD, the second most abundant constituent of cannabis
   
inverse agonist [9]. It activates the TRPV1 channel [10] and 5-HT1A receptors [11]. It also is a GPR55 receptor antagonist [12] and inhibits
the enzymatic activity of fatty acid amide hydrolase (FAAH) and the endocannabinoid anandamide’s reuptake [13].
CBG has recently been evaluated in vitro and in vivo models [14]. It is a precursor of the most abundant cannabinoids and expresses an

    

PD [18], and Huntington’s disease [19].
A recently published general patient survey of CBG-predominant cannabis use reported improvement in anxiety, chronic pain, depres-

very few adverse side-effects [20].
Parkinson’s Disease and Dementia with Lewy Bodies, Patients Under Treatment with Standardized Extracts Enriched in
Cannabidiol and Cannabigerol: Descriptive Observations in Preparation for a Phase 2a Clinical Trial
02
Citation: Flávio Henrique de Rezende Costa., et al. “Parkinson’s Disease and Dementia with Lewy Bodies, Patients Under Treatment with
Standardized Extracts Enriched in Cannabidiol and Cannabigerol: Descriptive Observations in Preparation for a Phase 2a Clinical Trial”.
EC Neurology 14.5 (2022): 01-15.
 
(Epidiolex) [22] have been medically approved for the management of pain/spasticity in multiple sclerosis and for refractory epilepsy
in Dravet/Lennox Gastaut syndromes.
In 2019 the National Health Surveillance Agency (ANVISA) in Brazil created a protocol for the registration of CPGF [23]. Only formu-
  -
      
Cannabis
formulations can also be imported legally to Brazil, provided that they have received sanitary approval in the country of origin [25].
Clinical studies have demonstrated the potential of CBD in improving quality of life [26], reducing psychotic symptoms [27], and at-
tenuating REM sleep behavioral disorder [28]. The high density of CB1 cannabinoid receptors in the basal ganglia also led to its investiga-

levodopa-induced dyskinesias in one small clinical trial [30] but most of the few available studies have shown a limited role for phytocan-
nabinoids with regard to improvement of motor symptoms in PD [31].
Recent clinical and preclinical evidence has suggested that CBD may have antipsychotic properties [32]. In contrast to some of the
available anti-psychotic drugs CBD also appears to have no cardiac toxicity and does not increase the QT interval [33].
This paper describes pilot descriptive data collected in preparation for a controlled phase 2a study and describes the response of a
small number of patients with PD and DLB patients treated with four different CBD or CBD/CBG formulations.
-
SA. The extracts are produced in Florida-USA under GMP compliance (CBCeuticals Laboratories/Health Meds Laboratories) and imported
-
tion of analysis for each batch and passed the stability test protocol ASTM-F1980.
Methods
The medical records of 14 PD and 5 DLB patients were reviewed between April 2020 and June 2021. All patients were followed up in
a single medical facility (Multidisciplinary Clinic- Botafogo- Rio de Janeiro/Brazil).
A neurologist (FHRC), specialized in movement disorders, established a PD diagnosis based on the Queen Square Brain Bank Criteria
[34] and DBL using the Movement Disorders Society’s criteria [35]. At the baseline clinical interview the main complaints were divided
into motor or non-motor categories. The most frequent unmet needs for PD were disruptive RBD (N = 10), anxiety (n = 5), insomnia (n =

For DLB: disorientation with getting lost (n = 4), disruptive RBD (3), sleep fragmentation (n = 5), night agitation (n = 3), severe/complex
hallucinations (n = 4), and delirium (5).
           
(Health Meds Laboratories-Brazil) were available for prescription as enriched standardized extracts: 1) Cannabidiol (CBD) broad spec-
trum (100 mg/mL) 2) Cannabidiol/Cannabigerol (CBD/CBG) (2:1) broad spectrum (100mg/mL) (3) Cannabidiol/Cannabigerol (CBD/
CBG) (2:1) +THC 0.3% full spectrum (100mg/mL) 4) Cannabidiol (CBD)+THC 0.3% full spectrum (100mg/mL). All CBD or CBD/CBG for-

Parkinson’s Disease and Dementia with Lewy Bodies, Patients Under Treatment with Standardized Extracts Enriched in
Cannabidiol and Cannabigerol: Descriptive Observations in Preparation for a Phase 2a Clinical Trial
03
Citation: Flávio Henrique de Rezende Costa., et al. “Parkinson’s Disease and Dementia with Lewy Bodies, Patients Under Treatment with
Standardized Extracts Enriched in Cannabidiol and Cannabigerol: Descriptive Observations in Preparation for a Phase 2a Clinical Trial”.
EC Neurology 14.5 (2022): 01-15.
The formulations were presented in opaque bottles containing 60 ml. The excipient was Organic Medium Chain Triglycerides (MCT) oil
(USP - FDA GRAS). A graduated micropipette was available in each of the bottles, protected by a child-proof cap. Each drop of the solution
had 2.5 mg CBD or 2.5 mg CBD / CBG (2:1); therefore, 40 drops (1ml) contained 100mg of CBD or 100mg of CBD/CBG (2:1). In THC 0.3%
formulations each 1 mL delivered 3 mg of THC.
 
the concept of “start low and go slow” (2.5 mg - 12.5 mg CBD at bedtime) with a focus on determining the lowest dose for symptomatic
control. The dose titration was variable, depending on the target symptoms.
A pharmacist oversaw pharmacovigilance by phone or by email contact.
Follow-up medical consultations took place every 2-3 months when target symptoms were reassessed, and a physician made dosage
adjustments.
Ethical considerations
The study received ethics committee approval and institutional board review (CEP/UNESA: 45936121.2.0000.5284 / Plataforma Bra-
sil). All patients received the prescriptions based on ANVISA resolution number 335 - which regulates the importing process of cannabis
derivatives to Brazil (25). Patients signed an informed consent form and completed an electronic form made available on the regulator’s

the prescribing physician’s name, the formulation, and the patient’s name associated with a code generated by the regulatory agency. The
laboratory staff could assist the patients or their guardians during the entire ANVISA clearance process. All personal data followed the
guidelines of the Federal Government’s data protection law.
Measurements and calculations
Data were extracted from the records of each patient (ComAmigo® version 9.16.61 www.cebim.com.br). The variables age, gender,
disease duration, follow-up duration, H/Y stage, levodopa, and cannabinoids doses were tabulated in the SPSS (version 18 for MAC) for
calculating the means and standard deviations (SD). The concomitant use of psychotropic medications was noted in each case.

LIMITED, or INEFFECTIVE. Symptoms graded as GOOD/MODERATE were deemed satisfactory, and LIMITED/INEFFECTIVE were con-
sidered unsatisfactory. Changes in drug prescription regimes were noted, focusing on recording reduction or withdrawal of sedatives or
neuroleptics.
Data was sent to a medical auditor (ELT) and another neurologist specializing in movement disorders (MS) to check and discuss any
inconsistencies.
Results
The mean age of the PD patients was 72.6 years (42 - 94) and for DLB , 82.2 years. (8 3- 92). Most of the patients were male (PD =
10 and DLB = 3). The disease duration was 6.57 years. (± 2.70) for PD and 4.2 years. (± 1.30) for DLB. The follow-up duration was 3.78
months (± 5.63) for PD and 4.2 months (± 1.30) for DLB. The average levodopa dose was 490 mg (± 212), with a mean H / Y of 3 (± 0.84)
in the PD group (Table 1 to 6).
Parkinson’s Disease and Dementia with Lewy Bodies, Patients Under Treatment with Standardized Extracts Enriched in
Cannabidiol and Cannabigerol: Descriptive Observations in Preparation for a Phase 2a Clinical Trial
04
Citation: Flávio Henrique de Rezende Costa., et al. “Parkinson’s Disease and Dementia with Lewy Bodies, Patients Under Treatment with
Standardized Extracts Enriched in Cannabidiol and Cannabigerol: Descriptive Observations in Preparation for a Phase 2a Clinical Trial”.
EC Neurology 14.5 (2022): 01-15.
Parkinson’s Disease and Dementia with Lewy Bodies, Patients Under Treatment with Standardized Extracts Enriched in
Cannabidiol and Cannabigerol: Descriptive Observations in Preparation for a Phase 2a Clinical Trial
05
PD DLB
Patients (n) 14 5
Age (years) 72.6 (42-94) 82.2 (83-92)
Gender n (%) (male) 10 (72) 3 (60)
Disease Duration (yrs) (SD) 6.57 (± 2.70) 4.2 (± 1.30)
Follow up duration (months) (SD) 3.78 (± 5.63) 4.2 (± 1.30)
Hoehn and Yahr (SD) 3 (± 0.84)
Mean Levodopa dose mg (SD) 490 (± 212)
Daily CBD mean dose mg (SD) 67.50 (± 38.6) 52 (± 44.8)
Daily CBG mean dose mg (SD) 22.50 (± 16.82) 8.75 (± 8.33)
Daily THC mean dose mg (SD)* 2.32 (± 1.46) 0.02 mg (± 0.01)
Daily THC mean dose mg (SD)** 0.83 (± 2.24) 0.02 mg (± 0.01)
Table 1: Patients’ characteristics.
  
with Lewy bodies. * n=5 PD and n=1 DLB patients on THC containing formulations. ** All patients included.
# Diagnosis Gender Age Disease
Duration
(years)
Disease Stage H/Y Comorbidities Initial
Concurrent
Drugs
#1 PD M 63 6 6 2 Essential hyperten-
sion, Chronic pain
(coxofemural osteo-
arthritis)
Levodopa 600mg/day;
Rasagiline 1mg/day,
domperidone 10mg t.i.d,
Amantadine 100mg t.i.d,
Rotigotine 8mg/day,
#2 PD M 69 5 1 3 Obesity Levodopa 900mg/day,
Rasagiline 1mg/day, entaca-
pone 1 gr/dia, clonazepam
2mg, duloxetin 60mg, dom-
peridone 10mg t.i.d
#3 PD F 88 7 5 3 Essential hyperten-
sion, osteoarthritis
(low back pain)
Levodopa 400mg/day,
sertraline 100mg//day,
zolpidem 10mg bedtime
#4 PD M 46 6 5 2 Levodopa 600mg/day;
Rasagiline 1mg/day,
domperidone 10mg t.i.d,
pramipexol 4.5 mg/day
#5 PD M 53 6 1 3 Levodopa 500mg/day;
Rasagiline 1mg/day,
domperidone 10mg t.i.d,
clozapine 12.5mg/day
Citation: Flávio Henrique de Rezende Costa., et al. “Parkinson’s Disease and Dementia with Lewy Bodies, Patients Under Treatment with
Standardized Extracts Enriched in Cannabidiol and Cannabigerol: Descriptive Observations in Preparation for a Phase 2a Clinical Trial”.
EC Neurology 14.5 (2022): 01-15.
Parkinson’s Disease and Dementia with Lewy Bodies, Patients Under Treatment with Standardized Extracts Enriched in
Cannabidiol and Cannabigerol: Descriptive Observations in Preparation for a Phase 2a Clinical Trial
06
#6 PD M 80 12 6 4 PD Dementia (PIGD) Levodopa 400mg/day,
Rasagiline 1mg/day, Riv-
astigmine Patch 9,5mg/day,
melatonin 5mg
#7 PD M 94 8 3 4 Osteoarthritis Levodopa 400mg/day,
Rasagiline 1mg/day
#8 PD M 77 6 4 2.5 Essential hyperten-
sion
Levodopa 300mg/day, Riv-
astigmine Patch 9,5mg/day
#9 PD M 90 12 1 5 Advanced pharyn-
geal neoplasm (pal-
liative care)
Levodopa 400mg/day,
Rasagiline 1mg/day, Riv-
astigmine Patch 9,5mg/day,
melatonin 5mg, escitalo-
pram 5mg/day, clonazepam
0.25mg
#10 PD M 85 6 2 3 Colostomy (past
bowel diverticulum
perforation)
Levodopa 900mg/day,
amantadine 100mg/day,
venlafaxine 75mg/day,
Lithium (prescribed by
another physician)
#11 PD F 88 4 5 3 Levodopa 500mg/day;
Rasagiline 1mg/day,
amantadine 100mg/day,
Lorazepan 2mg bedtime
#12 PD F 79 2 5 2 Hypothyroidism=
levothyroxine
125mcg/day
Levodopa 150mg/day, rasa-
giline 1mg/day, domperi-
-
tisone 0.2 mg bedtime
#13 PD M 80 5 5 3 Levodopa 300mg/dia,
Rotigotine 4mg/day, galan-
tamine 16mg/day, zolpidem
6.25mg, quetiapine 75mg
bedtime,
#14 PD F 75 7 4 3 Thrombophilia Levodopa 500mg/day;
Rasagiline 1mg/day, Rotigo-
tine 8mg/day, amantadine
100mg/day, venalafaxine
75mg/day, domperidone
30mg/day, dabigatran
Table 2: PD: demographics, comorbidities, and initial concurrent treatment.
PIGD=Postural instability with gait disability. PD= Parkinson’s disease.
Citation: Flávio Henrique de Rezende Costa., et al. “Parkinson’s Disease and Dementia with Lewy Bodies, Patients Under Treatment with
Standardized Extracts Enriched in Cannabidiol and Cannabigerol: Descriptive Observations in Preparation for a Phase 2a Clinical Trial”.
EC Neurology 14.5 (2022): 01-15.
Parkinson’s Disease and Dementia with Lewy Bodies, Patients Under Treatment with Standardized Extracts Enriched in
Cannabidiol and Cannabigerol: Descriptive Observations in Preparation for a Phase 2a Clinical Trial
07
Number Diagnosis Formulation Daily CBD Dose
(mg)
Daily CBG
Dose(mg)
Daily THC
Dose(mg)
#1 PD Cannabidiol + Cannabigerol 2:1 (100mg/mL) + THC 0.3% 100 50 4,5
#2 PD Cannabidiol (100mg/mL) 100
#3 PD Cannabidiol + Cannabigerol 2:1 (100mg/mL) 41.66 20.83
#4 PD Cannabidiol (100mg/mL) + THC 0.3 % 25 0,75
#5 PD Cannabidiol + Cannabigerol 2:1 (100mg/mL) 12.5
#6 PD Cannabidiol (100mg/mL) 50
#7 PD Cannabidiol + Cannabigerol 2:1 (100mg/mL) 8.33 4.16
#8 PD Cannabidiol (100mg/mL) 125 0
#9 PD Cannabidiol + Cannabigerol 2:1 (100mg/mL) + THC 0.3% 33.33 16.66 1,5
#10 PD Cannabidiol (100mg/mL) 75 0
#11 PD Cannabidiol + Cannabigerol 2:1 (100mg/mL) + THC 0.3% 41.66 20.83 1,87
#12 PD Cannabidiol (100mg/mL) + THC 0.3 % 100 3
#13 PD Cannabidiol (100mg/mL) 100
#14 PD Cannabidiol (100mg/mL) 100
#1 DLB Cannabidiol (100mg/mL) 100
#2 DLB Cannabidiol (100mg/mL) 100
#3 DLB Cannabidiol + Cannabigerol 2:1 (100mg/mL) 30 15
#4 DLB Cannabidiol + Cannabigerol 2:1 (100mg/mL) + THC 0.3% 25 2.5 0.025
#5 DLB Cannabidiol (100mg/mL) 5
Table 3: Formulations and daily doses of each patient.
# Diagnosis Unmet Need Before Treatment Effect On Un-
met Needs
Overall Ef-
fect
Comments
#1 PD 1)Pain**** and compulsion 2)
insomnia 3) RBD* 4) Anxiety***
GOOD GOOD Rigidity reduction only on 4.5 mg of THC
#2 PD 
gait) 2) Low back pain 3) RBD
INEFFECTIVE LIMITED The patient discontinued treatment after
one month. Improvement in sleep duration.
#3 PD 1) insomnia 2) RDB* 3) pain 4)
Mild hallucinations (benign)**
GOOD GOOD Zolpidem withdrawn. Fewer episodes of
somnambulism.
#4 PD 1) Problematic wearing off phe-
nomenon 2) impulsivity (compul-
sive internet stock market “day
trade” behavior 3) severe anxiety
4) RBD*/insomnia
MODERATE LIMITED No impact on motor symptoms / no impact
on wearing off/ Drowsiness in doses
>50mg CBD/THC 0.3%. Patient on prepara-
tion for DBS.
Citation: Flávio Henrique de Rezende Costa., et al. “Parkinson’s Disease and Dementia with Lewy Bodies, Patients Under Treatment with
Standardized Extracts Enriched in Cannabidiol and Cannabigerol: Descriptive Observations in Preparation for a Phase 2a Clinical Trial”.
EC Neurology 14.5 (2022): 01-15.
Parkinson’s Disease and Dementia with Lewy Bodies, Patients Under Treatment with Standardized Extracts Enriched in
Cannabidiol and Cannabigerol: Descriptive Observations in Preparation for a Phase 2a Clinical Trial
08
#5 PD 1) Problematic motor and “non-
motor wearing-off phenomenon.
Severe tremor 2) Transient
psychosis/hallucinations (induced
by dopaminergic overstimulation:
suicide attempt
LIMITED LIMITED Clozapine withdrawal. No effect on the

preparation for DBS.
#6 PD 1) Hallucinations** 2) cognitive
decline 3) RDB*/ wandering 4)
severe gait disability
GOOD GOOD Remains without the use of neuroleptics.
No impact on motor symptoms. Add on ef-
fect on hallucinations
#7 PD 1) Insomnia 2) Nocturnal pain ****
-
tions (anxiety***) 4) RBD*
GOOD GOOD Market attenuation of non-motor nocturnal

#8 PD 1) Anxiety*** 2) Complex halluci-
nations** 3) Insomnia 4) RBD*
GOOD GOOD Stable without neuroleptics. No impact on
motor symptoms
#9 PD 1) Hallucinations** 2) Cogni-
tive decline 3) Severe rigidity 4)
Insomnia
GOOD MODERATE Palliative care in end of life. No effect on
rigidity. Morphine sparing effect. No end-
of-life delirium. Patient died comfortably
#10 PD 1)Gait balance problems 2) Apathy
3) Mild depressive symptoms
GOOD GOOD Better on the balance after lithium with-
dr aw.
#11 PD 1) Severe pain****: Dopamine
agonist-induced cervical dystonia
2) anxiety*** 3) RBInsomniania
GOOD GOOD Other actions for pain management: imme-
diate pramipexol withdrawal and US-guid-
ed botulinum toxin cervical injection.
#12 PD 1) Severe levodopa/rasagiline
induced nausea and hypotension
2) Anxiety
GOOD GOOD Nausea improvement. Orthostatic hypoten-

#13 PD 1) Moderate hallucinations**2)
RBD* and insomnia 3) Cognitive
decline (PDD)
GOOD GOOD Quetiapine dose unchanged. Add on effect
on hallucinations.
#14 PD 1) wearing off (freezing of gait) 2)
Cognitive decline 3) RBD*/ benign
hallucinations** 5) Anxiety***
(despite venlafaxine use) 6) pain
(knee arthrosis)
GOOD MODERATE No impact on wearing off phenomenon
RDB symptom abolished/marked improve-
ment of hallucinations
Table 4: RBD= Rem sleep behavior disorder. *RBD symptoms abolished and substantial increment in sleep duration. ** Marked improve-
ment in hallucinations. *** Marked response in anxiety symptoms. **** Pain responders. PD= Parkinson’s disease.
Citation: Flávio Henrique de Rezende Costa., et al. “Parkinson’s Disease and Dementia with Lewy Bodies, Patients Under Treatment with
Standardized Extracts Enriched in Cannabidiol and Cannabigerol: Descriptive Observations in Preparation for a Phase 2a Clinical Trial”.
EC Neurology 14.5 (2022): 01-15.
Parkinson’s Disease and Dementia with Lewy Bodies, Patients Under Treatment with Standardized Extracts Enriched in
Cannabidiol and Cannabigerol: Descriptive Observations in Preparation for a Phase 2a Clinical Trial
09
# Diag-
nosis
Gender Age Disease
Duration
(yrs)
Disease Stage H/Y Comorbidi-
ties
Initial Concurrent
Drugs
#1 DLB M 83 3 2 N/A Coronary disease (past
coronary angioplasty with
stents implants)
Levodopa 400mg/day,
melatonin 10mg bedtime,
rivastigmine Patch 4,5 mg/day,
eszopiclone 2mg bedtime
#2 DLB F 71 3 5 N/A Rivastigmine Patch 9,5 mg/
day, melatonin 5mg day,
memantine 10 mg bedtime,
mirtazapine 15mg bedtime,
alprazolam 0.5mg bedtime
#3 DLB M 87 4 4 N/A 
vein thrombosis / parox-

anticoagulant: dabigatran
Donepezil 10mg/day, meman-
tine 20mg/day, escitalopram
10mg/day, alprazolam 2mg/
day, levodopa 400mg/day
#4 DLB F 78 6 5 N/A Severe weight loss Rivastigmine Patch 4,5 mg/
day, trazodone 25mg/day,
olanzapine 2,5mg, eszopiclone
2mg/day
#5 DLB M 92 5 5 N/A Arterial peripheral vas-
cular disease, Pulmonary

tract infections with de-
lirium (prolonged double J
catheter)
Rivastigmine Patch 13.3 mg/
day, clozapine 12.5mg bedtime,
venlafaxine 75mg/day
Table 5: DLB: demographics, comorbidities, and initial concurrent treatment. DLB= Dementia with Lewy bodies.
# Diagnosis Unmet need before treatment Effect on unmet needs Overall Effect comments
#1 DLB -
ing, agitation, and RBD 3) Severe complex
hallucinations, 4) Drug hypersensitivity.
INEFFECTIVE INEFFECTIVE 
#2 DLB 1) Severe complex hallucinations*** 2) wan-
dering** 3) anxiety
MODERATE LIMITED Limited effect on the
speed of cognitive
decline. The patient still
deteriorating on multiple
other aspects: Language
visuospatial skills
Citation: Flávio Henrique de Rezende Costa., et al. “Parkinson’s Disease and Dementia with Lewy Bodies, Patients Under Treatment with
Standardized Extracts Enriched in Cannabidiol and Cannabigerol: Descriptive Observations in Preparation for a Phase 2a Clinical Trial”.
EC Neurology 14.5 (2022): 01-15.
#3 DLB 1) Severe hallucinations*** 2) wander-
ing** 3) RBD* 4) fast cognitive decline and
sundowning
GOOD MODERATE Mild mental confusion
/ somnolence in doses
above 45mg of CBD/CBG
2:1. Stable on the actual
dose.
#4 DLB 1) Severe hallucinations*** 2) wandering**
3) RBD*, fast cognitive decline and sun-
downing 4) severe parkinsonism 5) severe
weight loss
GOOD GOOD Olanzapine and eszopi-
clone was withdrawn
#5 DLB 1) Insomnia and nocturnal agitation 2)
Marked parkinsonism with gait balance


GOOD MODERATE Clozapine was with-
drawn
Table 6: *RBD responders. ** Marked improvement in wandering. *** Marked response in hallucinations.
DLB= Dementia with Lewy bodies.
Parkinson’s Disease and Dementia with Lewy Bodies, Patients Under Treatment with Standardized Extracts Enriched in
Cannabidiol and Cannabigerol: Descriptive Observations in Preparation for a Phase 2a Clinical Trial
10
Regarding prescribed formulations, PD patients were on CBD enriched n = 6, CBD/CBG 2:1 enriched n = 3, CBD/CBG 2:1 enriched +
THC 0.3% n = 3, CBD/THC 0.3% n = 2 full spectrum. DBL patients were on CBD enriched n = 3, CBD/CBG 2: 1 enriched n = 1, CBD/CBG 2:
1 enriched +THC 0.3% n = 1.
The average CBD dose was 67.5 mg for PD (± 38.6) and 52mg (± 44.8) for DLB. The CBG dose was 22.5 mg for PD (± 16.82) and 8.7mg
(± 8.33) for DLB. The average THC dose was 2.32 mg for PD (± 1.46) and 0.02 mg for DLB (± 0.01); this variable was calculated only in
patients using THC-containing formulations. Considering all patients in the PD and DLB groups, the average THC dose was 0.83 mg (±
2.24) for PD, and 0.025 mg (± 0.01) for DLB.
Marked improvements were observed on RBD (9/10), insomnia (9/9), anxiety (5/5) in the PD patients. There was also improvement
in reported pain (3/5), impulsive-compulsive behaviors (ICB’s) (2/2), and visual hallucinations (6/7). All pain responders were on for-
mulations with CBG and THC.
RBD symptoms were markedly reduced in 9 patients (PD#1, PD#3, PD#4, PD#6, PD#7, PD#8, PD#11, PD#13, PD#14). For instance,
PD # 14 had a long history of sleep disturbance having kicked her husband several times during vivid dreams episodes. All her symptoms
vanished after 50mg CBD nocte. The dose was titrated to 50mg twice a day, and this patient also experienced a marked attenuation of late
afternoon visual hallucinations.

(PD#1, PD#7, PD#8, PD#11, PD#14). PD#11 had experienced several months of pain and antecollis dystonic posture. The patient was in
great distress, markedly anxious, and experiencing severe insomnia. Dopamine agonist-induced cervical dystonia was initially considered
as a cause of pain and abnormal cervical posture. Pramipexole was suspended and rasagiline and low dose amantadine were slowly intro-
duced but with no improvement. After four weeks, CBD/CBG 2:1 + THC 0.3% formulation was started and titrated to 62.5 mg (25 drops)
at night. A marked improvement in anxiety symptoms and sleep disturbance was promptly observed.
Citation: Flávio Henrique de Rezende Costa., et al. “Parkinson’s Disease and Dementia with Lewy Bodies, Patients Under Treatment with
Standardized Extracts Enriched in Cannabidiol and Cannabigerol: Descriptive Observations in Preparation for a Phase 2a Clinical Trial”.
EC Neurology 14.5 (2022): 01-15.
Parkinson’s Disease and Dementia with Lewy Bodies, Patients Under Treatment with Standardized Extracts Enriched in
Cannabidiol and Cannabigerol: Descriptive Observations in Preparation for a Phase 2a Clinical Trial
11
A modest response in musculoskeletal pain was observed only in those patients with formulations that contained CBD or CBG with or
without THC (PD#1, PD #7, and PD#11). CBD isolated formulations did not produce analgesic effects in PD#2 and PD#14.
An antipsychotic effect occurred in six patients (PD#3, PD#6, PD#8, PD#9, PD#13, PD#14)- usually associated with improved sleep
quality and decreased anxiety symptoms.
In DLB patients, improvements in wandering (3/4), disruptive RBD (2/3), sleep fragmentation (4/5), night agitation (2/3), and hallu-
cinations (4/5) were reported and in two patients (DLB#4 and DLB#5) with delirium and agitation it was possible to discontinue sedative
and anti-psychotic medication. DLB#3 showed a marked reduction in visual hallucinations, despite having drowsiness with CBD/CBG 2:

and no visual hallucinations at the current dose.
Concerning DLB#1, intervention with CBD enriched (THC Free) was considered ineffective. The patient was looked after by his elderly
-

No improvement in motor symptoms was observed, except in patient #1 (CBD/CBG 2:1 + THC 0.3%) who reported reduced stiffness
with 4.5mg THC daily. PD #5 had severe off tremor that failed to respond This patient is currently undergoing a evaluation for a DBS
surgical implant.
Discussion
This study provides tentative support for earlier studies demonstrating phytocannabinoids’ potential in managing some PD non-

The high average age in both groups, multiple comorbidities, concurrent use of multiple drugs, and relatively long disease duration in

The response rate was high in sleep disorders, especially in the disruptive forms of RBD and PD/DLB sleep fragmentation. In PD, 9 of
-
derline RBD and sleep disruption both in PD and DLB [36]. Preclinical studies have shown that CBD enhances in vivo acetylcholine levels
from the basal forebrain and increments sleep duration [37].
Current RBD drug treatment options are melatonin and clonazepam [38] - the latter although highly effective can interfere with cogni-
tion and normal sleep architecture, and put patients at greater risk of falls [39].
-
ety attacks) and improved sleep quality. Mild drowsiness occurred, however, in CBD/CBD (2:1) dose above 12.5 mg (5 drops)- indicating
that dose titration should be individualized and symptom-focused.

-
Citation: Flávio Henrique de Rezende Costa., et al. “Parkinson’s Disease and Dementia with Lewy Bodies, Patients Under Treatment with
Standardized Extracts Enriched in Cannabidiol and Cannabigerol: Descriptive Observations in Preparation for a Phase 2a Clinical Trial”.
EC Neurology 14.5 (2022): 01-15.
Parkinson’s Disease and Dementia with Lewy Bodies, Patients Under Treatment with Standardized Extracts Enriched in
Cannabidiol and Cannabigerol: Descriptive Observations in Preparation for a Phase 2a Clinical Trial
12
sive painting), and reduced muscle stiffness (need a reference here). Previous studies have also shown that doses of isolated CBD between

Our study emphasizes the need for lower doses in all formulations, compared with previous reports in the elderly, and provides some
support for cannabinoids exerting an entourage effect [40].
   
dementia and increased mortality in PD patients [41]. Current treatment options are restricted to cholinomimetics and atypical antipsy-
chotics such as clozapine In DBL, the use of neuroleptics is even more challenging due to neuroleptics hypersensitivity and neuroleptics
have been associated with increased mortality in dementias [39].
A moderate response to pain was observed only in those on CBG or THC formulations (PD#1, PD#7, PD#11), supporting the view that
combinations of cannabinoids may be best for this indication [40]. .
  
promptly reversed with dose reduction.
Even though the data were structured for each patient, no previously validated scale was applied in this study and there was no ran-
domization. The outcomes were assessed exclusively based on the patients’ or caregivers’ reports in a semi-structured clinical interview
conducted by a neurologist with training in movement disorders. On the other hand, checking the data by an independent neurologist and
a medical auditor helped with data transparency.
Finally, the present work was possible due to Brazilian legislation changes. The recent ANVISA regulations provided access to stan-
dardized extracts via import process, although not yet available in the Brazilian National Health System under universal coverage [25].
  -
cacy in various medical indications [23].
Perspectives for Future Studies
            
encourage the need for well-conducted randomized clinical trials with CBD or CBD/CBG to examine its antipsychotic potential. Further
trials with phytocannabinoids for tremor and l-dopa induced dyskinesias in PD should also be conducted.
Conclusions
This report describes the potential of CBD/CBG formulations for non-motor symptoms treatment in PD and DLB- such as anxiety, sleep
disorders, visual hallucinations, impulsivity, and pain and will be helpful in designing a phase 2a randomized placebo-controlled clinical
trial.

Prof. Flávio Rezende works part-time in Research and Development (R&D) for Health Meds Laboratories (Brazil) and advisor physician
for Cbeuticals Laboratories (USA).
Simone Pellegrino de Oliveira works full-time as a Pharmacist for Health Meds-Brazil.
Citation: Flávio Henrique de Rezende Costa., et al. “Parkinson’s Disease and Dementia with Lewy Bodies, Patients Under Treatment with
Standardized Extracts Enriched in Cannabidiol and Cannabigerol: Descriptive Observations in Preparation for a Phase 2a Clinical Trial”.
EC Neurology 14.5 (2022): 01-15.
13
Parkinson’s Disease and Dementia with Lewy Bodies, Patients Under Treatment with Standardized Extracts Enriched in
Cannabidiol and Cannabigerol: Descriptive Observations in Preparation for a Phase 2a Clinical Trial
1. Goetz CG. “The History of Parkinson’s Disease: Early Clinical Descriptions and Neurological Therapies”. Cold Spring Harbor Perspec-
tives in Medicine 1.1 (2011): a008862-a008862.
2. Russo EB. “History of cannabis and its preparations in saga, science, and sobriquet”. Chemistry and Biodiversity 4.8 (2007): 1614-
1648.
3. Zuardi. “History of cannabis as a medicine: a review”. The Revista Brasileira de Psiquiatria (2006).
4. Gaoni Y and Mechoulam R. “Isolation, Structure, and Partial Synthesis of an Active Constituent of Hashish”. Journal of the American
Chemical Society 86.8 (1964): 1646-1647.
5. Mechoulam R. “Marihuana chemistry”. Science 168.3936 (1970): 1159-1166.
6. Matsuda LA., et al. “Structure of a cannabinoid receptor and functional expression of the cloned cDNA”. Nature 346.6284 (1990):
561-564.
7. Munro S., et al. “Molecular characterization of a peripheral receptor for cannabinoids”. Nature 365.6441 (1993): 61-65.
8. Cristino L., et al. “Cannabinoids and the expanded endocannabinoid system in neurological disorders”. Nature Reviews Neurology 16.1
(2020): 9-29.
9. McPartland JM., et al-
tematic review”. British Journal of Pharmacology 172 (2015): 737-753.
10. De Petrocellis L., et al. “Effects of cannabinoids and cannabinoid-enriched Cannabis extracts on TRP channels and endocannabinoid
metabolic enzymes”. British Journal of Pharmacology 163.7 (2011): 1479-1494.
11. Russo EB., et al. “Agonistic properties of cannabidiol at 5-HT1a receptors”. Neurochemical Research 30.8 (2005): 1037-1043.
12. Rylander D., et al. “Pharmacological modulation of glutamate transmission in a rat model of L-DOPA-induced dyskinesia: Effects on
motor behavior and striatal nuclear signaling”. Journal of Pharmacology and Experimental Therapeutics 330.1 (2009): 227-235.
13. Leweke FM., et al. “Cannabidiol enhances anandamide signaling and alleviates psychotic symptoms of schizophrenia”. Translational
Psychiatry 2.3 (2012).
Bibliography
Eduardo Rydz works full-time as Director of Access for Health Meds-Brazil.
Dr. Gabriel de Castro Micheli works part-time as an MSL (Medical Science Liaison) for Health Meds Laboratories.
Jaron Gladstone is CEO and Founder of CBCeuticals Laboratories- Coral Springs- Florida- United States.
Brian Ebner is V.P of production at CBCeuticals Laboratories – Coral Springs- Florida- United States.



Citation: Flávio Henrique de Rezende Costa., et al. “Parkinson’s Disease and Dementia with Lewy Bodies, Patients Under Treatment with
Standardized Extracts Enriched in Cannabidiol and Cannabigerol: Descriptive Observations in Preparation for a Phase 2a Clinical Trial”.
EC Neurology 14.5 (2022): 01-15.
14
Parkinson’s Disease and Dementia with Lewy Bodies, Patients Under Treatment with Standardized Extracts Enriched in
Cannabidiol and Cannabigerol: Descriptive Observations in Preparation for a Phase 2a Clinical Trial
14. Nachnani R., et al. “The Pharmacological Case For Cannabigerol (Cbg)”. Journal of Pharmacology and Experimental Therapeutics
(2020).
15. O’Sullivan SE. “An update on PPAR activation by cannabinoids”. British Journal of Pharmacology 173 (2016): 1899-1910.
16. Ryz NR., et alCannabis and Can-
nabinoid Research 2.1 (2017): 210-216.
17. Ligresti A., et al. “Antitumor activity of plant cannabinoids with emphasis on the effect of cannabidiol on human breast carcinoma”.
Journal of Pharmacology and Experimental Therapeutics 318.3 (2006): 1375-1387.
18. García C., et al    
-
tion 15.1 (2018).
19. Díaz-Alonso J., et al. “VCE-003.2, a novel cannabigerol derivative, enhances neuronal progenitor cell survival and alleviates symptom-
atology in murine models of Huntington’s disease (2016): 6.
20. Russo EB., et al. “Survey of Patients Employing Cannabigerol-Predominant Cannabis Preparations: Perceived Medical Effects, Ad-
verse Events, and Withdrawal Symptoms”. Cannabis and Cannabinoid Research (2021).
21. Perras C. “Sativex for the management of multiple sclerosis symptoms”. Issues in Emerging Health Technologies 72 (2005): 1-4.
22. Devinsky O., et al. “Trial of Cannabidiol for Drug-Resistant Seizures in the Dravet Syndrome”. The New England Journal of Medicine
376.21 (2017): 2011-2020.
23. ANVISA. RDC 327 (2019).
24. ANVISA. RDC 318 (2019).
25. ANVISA. RDC 335 (2020).
26. MH C., et al. “Effects of Cannabidiol in the Treatment of Patients With Parkinson’s Disease: An Exploratory Double-Blind Trial”. Journal
of Psychopharmacology 28.11 (2014).
27. Zuardi AW., et al. “Cannabidiol for the treatment of psychosis in Parkinson’s disease”. Journal of Psychopharmacology 23.8 (2009):
979-983.
28. Chagas MHN., et al. “Cannabidiol can improve complex sleep-related behaviours associated with rapid eye movement sleep behav-
iour disorder in Parkinson’s disease patients: a case series”. Journal of Clinical Pharmacy and Therapeutics 39.5 (2014): 564-566.
29. Frankel JP., et al. “Marijuana for Parkinsonian tremor”. Journal of Neurology Neurosurgery and Psychiatry 53(1990): 436.
30. KA S., et al. “Cannabinoids Reduce Levodopa-Induced Dyskinesia in Parkinson’s Disease: A Pilot Study”. Neurology 57.11 (2001).
31. Crippa JAS., et alEuropean Archives of Psychiatry
and Clinical Neuroscience 269.1 (2019): 121-133.
32. McGuire P., et al. “Cannabidiol (CBD) as an Adjunctive Therapy in Schizophrenia: A Multicenter Randomized Controlled Trial”. The
American Journal of Psychiatry 175.3 (2018): 225-231.
Citation: Flávio Henrique de Rezende Costa., et al. “Parkinson’s Disease and Dementia with Lewy Bodies, Patients Under Treatment with
Standardized Extracts Enriched in Cannabidiol and Cannabigerol: Descriptive Observations in Preparation for a Phase 2a Clinical Trial”.
EC Neurology 14.5 (2022): 01-15.
15
Parkinson’s Disease and Dementia with Lewy Bodies, Patients Under Treatment with Standardized Extracts Enriched in
Cannabidiol and Cannabigerol: Descriptive Observations in Preparation for a Phase 2a Clinical Trial
33. Sellers EM., et al. “A Multiple-Dose, Randomized, Double-Blind, Placebo-Controlled, Parallel-Group QT/QTc Study to Evaluate the
Electrophysiologic Effects of THC/CBD Spray. Clinical Pharmacology in Drug Development 2.3 (2013): 285-294.
34. Hughes AJ., et al. “Accuracy of clinical diagnosis of idiopathic Parkinson’s disease: a clinico-pathological study of 100 cases”. Journal of
Neurology, Neurosurgery, and Psychiatry 55.3 (1992): 181-184.
35. Hughes AJ., et al. “Accuracy of clinical diagnosis of idiopathic Parkinson’s disease: a clinico-pathological study of 100 cases”. Journal of
Neurology, Neurosurgery, and Psychiatry 55.3 (1992): 181-184.
36. Viktor B., et al. “Pathological and clinical comparison of Parkinson’s disease dementia and dementia with Lewy bodies”. Orvosi Hetilap
161.18 (2020): 727-737.
37. Murillo-Rodríguez E., et al. “Systemic Injections of Cannabidiol Enhance Acetylcholine Levels from Basal Forebrain in Rats”. Neuro-
chemical Research 43.8 (2018): 1511-1518.
38. De Almeida CMO., et al. “Pharmacological treatment for REM sleep behavior disorder in Parkinson disease and related conditions: A
scoping review”. Journal of the Neurological Sciences (2018): 393.
39. Seppala LJ., et al. “Fall-Risk-Increasing Drugs: A Systematic Review and Meta-Analysis: II. Psychotropics”. Journal of the American
Medical Directors Association 19.4 (2018): 371.e11-371.e17.
40. Russo EB. “The Case for the Entourage Effect and Conventional Breeding of Clinical Cannabis: No and quot;Strain, and quot; No Gain”.
Frontiers in Plant Science 9 (2018): 1969.
41. Kempster PA., et al. “Relationships between age and late progression of Parkinson’s disease: a clinico-pathological study”. Brain 133.6
(2010): 1755-1762.
Volume 14 Issue 5 May 2022
© All rights reserved by Flávio Henrique de Rezende Costa., et al.
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Medical cannabis and individual cannabinoids, such as tetrahydrocannabinol (THC) and cannabidiol (CBD), are receiving growing attention in both the media and the scientific literature. The Cannabis plant, however, produces over 100 different cannabinoids, and cannabigerol (CBG) serves as the precursor molecule for the most abundant phytocannabinoids. CBG exhibits affinity and activity characteristics between THC and CBD at the cannabinoid receptors, but appears to be unique in its interactions with alpha-2 adrenoceptors and 5-HT1A Studies indicate that CBG may have therapeutic potential in treating neurological disorders (e.g., Huntington's Disease, Parkinson's Disease, and multiple sclerosis), inflammatory bowel disease, as well as having antibacterial activity. There is growing interest in the commercial use of this unregulated phytocannabinoid. This review focuses on the unique pharmacology of CBG, our current knowledge of its possible therapeutic utility, and its potential toxicological hazards. Significance Statement Cannabigerol (CBG) is currently being marketed as a dietary supplement and, as with cannabidiol (CBD) before, many claims are being made about its benefits. Unlike CBD, however, little research has been performed on this unregulated molecule, and much of what is known warrants further investigation to identify potential areas of therapeutic uses and hazards.
Article
Full-text available
Parkinson’s disease (PD) is a chronic neurodegenerative disorder characterized by motor symptoms such as bradykinesia, rest tremor, postural disturbances, and rigidity. PD is also characterized by non-motor symptoms such as sleep disturbances, cognitive deficits, and psychiatric disorders such as psychosis, depression, and anxiety. The pharmacological treatment for these symptoms is limited in efficacy and induce significant adverse reactions, highlighting the need for better treatment options. Cannabidiol (CBD) is a phytocannabinoid devoid of the euphoriant and cognitive effects of tetrahydrocannabinol, and preclinical and preliminary clinical studies suggest that this compound has therapeutic effect in non-motor symptoms of PD. In the present text, we review the clinical studies of cannabinoids in PD and the preclinical and clinical studies specifically on CBD. We found four randomized controlled trials (RCTs) involving the administration of agonists/antagonists of the cannabinoid 1 receptor, showing that these compounds were well tolerated, but only one study found positive results (reductions on levodopa-induced dyskinesia). We found seven preclinical models of PD using CBD, with six studies showing a neuroprotective effect of CBD. We found three trials involving CBD and PD: an open-label study, a case series, and an RCT. CBD was well tolerated, and all three studies reported significant therapeutic effects in non-motor symptoms (psychosis, rapid eye movement sleep behaviour disorder, daily activities, and stigma). However, sample sizes were small and CBD treatment was short (up to 6 weeks). Large-scale RCTs are needed to try to replicate these results and to assess the long-term safety of CBD.
Article
Full-text available
The topic of Cannabis curries controversy in every sphere of influence, whether politics, pharmacology, applied therapeutics or even botanical taxonomy. Debate as to the speciation of Cannabis, or a lack thereof, has swirled for more than 250 years. Because all Cannabis types are eminently capable of cross-breeding to produce fertile progeny, it is unlikely that any clear winner will emerge between the “lumpers” vs. “splitters” in this taxonomical debate. This is compounded by the profusion of Cannabis varieties available through the black market and even the developing legal market. While labeled “strains” in common parlance, this term is acceptable with respect to bacteria and viruses, but not among Plantae. Given that such factors as plant height and leaflet width do not distinguish one Cannabis plant from another and similar difficulties in defining terms in Cannabis, the only reasonable solution is to characterize them by their biochemical/pharmacological characteristics. Thus, it is best to refer to Cannabis types as chemical varieties, or “chemovars.” The current wave of excitement in Cannabis commerce has translated into a flurry of research on alternative sources, particularly yeasts, and complex systems for laboratory production have emerged, but these presuppose that single compounds are a desirable goal. Rather, the case for Cannabis synergy via the “entourage effect” is currently sufficiently strong as to suggest that one molecule is unlikely to match the therapeutic and even industrial potential of Cannabis itself as a phytochemical factory. The astounding plasticity of the Cannabis genome additionally obviates the need for genetic modification techniques.
Article
Full-text available
Background: Neuroprotection with cannabinoids in Parkinson's disease (PD) has been afforded predominantly with antioxidant or anti-inflammatory cannabinoids. In the present study, we investigated the anti-inflammatory and neuroprotective properties of VCE-003.2, a quinone derivative of the non-psychotrophic phytocannabinoid cannabigerol (CBG), which may derive its activity at the peroxisome proliferator-activated receptor-γ (PPARγ). The compound is also an antioxidant. Methods: We evaluated VCE-003.2 in an in vivo [mice subjected to unilateral intrastriatal injections of lipopolysaccharide (LPS)] model of PD, as well as in in vitro (LPS-exposed BV2 cells and M-213 cells treated with conditioned media generated from LPS-exposed BV2 cells) cellular models. The type of interaction of VCE-003.2 at the PPARγ receptor was furtherly investigated in bone marrow-derived human mesenchymal stem cells (MSCs) and sustained with transcriptional assays and in silico docking studies. Results: VCE-003.2 has no activity at the cannabinoid receptors, a fact that we confirmed in this study using competition studies. The administration of VCE-003.2 to LPS-lesioned mice attenuated the loss of tyrosine hydroxylase (TH)-containing nigrostriatal neurons and, in particular, the intense microgliosis provoked by LPS in the substantia nigra, measured by Iba-1/Cd68 immunostaining. The analysis by qPCR of proinflammatory mediators such as tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and inducible nitric oxide synthase (iNOS) in the striatum showed they were markedly elevated by the LPS lesion and strongly reduced by the treatment with VCE-003.2. The effects of VCE-003.2 in LPS-lesioned mice implied the activation of PPARγ receptors, as they were attenuated when VCE-003.2 was co-administered with the PPARγ inhibitor T0070907. We then moved to some in vitro approaches, first to confirm the anti-inflammatory profile of VCE-003.2 in cultured BV2 cells exposed to LPS. VCE-003.2 was able to attenuate the synthesis and release of TNF-α and IL-1β, as well as the induction of iNOS and cyclooxygenase-2 (COX-2) elicited by LPS in these cells. However, we found such effects were not reversed by GW9662, another classic PPARγ antagonist. Next, we investigated the neuroprotective effects of VCE-003.2 in cultured M-213 neuronal cells exposed to conditioned media generated from LPS-exposed cultured BV2 cells. VCE-003.2 reduced M-213 cell death, but again, such effects were not reversed by T0070907. Using docking analysis, we detected that VCE-003.2 binds both the canonical and the alternative binding sites in the PPARγ ligand-binding pocket (LBP). Functional assays further showed that T0070907 almost abolished PPARγ transcriptional activity induced by rosiglitazone (RGZ), but it did not affect the activity of VCE-003.2 in a Gal4-Luc system. However, T0070907 inhibited the effects of RGZ and VCE-003.2 on the expression of PPARγ-dependent genes upregulated in MSCs. Conclusions: We have demonstrated that VCE-003.2 is neuroprotective against inflammation-driven neuronal damage in an in vivo model of PD and in in vitro cellular models of neuroinflammation. Such effects might involve PPARγ receptors, although in silico and in vitro experiments strongly suggest that VCE-003.2 targets PPARγ by acting through two binding sites at the LBP, one that is sensitive to T0070907 (canonical binding site) and other that is not affected by this PPARγ antagonist (alternative binding site).
Article
Full-text available
Objective: Research in both animals and humans indicates that cannabidiol (CBD) has antipsychotic properties. The authors assessed the safety and effectiveness of CBD in patients with schizophrenia. Method: In an exploratory double-blind parallel-group trial, patients with schizophrenia were randomized in a 1:1 ratio to receive CBD (1000 mg/day; N=43) or placebo (N=45) alongside their existing antipsychotic medication. Participants were assessed before and after treatment using the Positive and Negative Syndrome Scale (PANSS), the Brief Assessment of Cognition in Schizophrenia (BACS), the Global Assessment of Functioning scale (GAF), and the improvement and severity scales of the Clinical Global Impressions Scale (CGI-I and CGI-S). Results: After 6 weeks of treatment, compared with the placebo group, the CBD group had lower levels of positive psychotic symptoms (PANSS: treatment difference=-1.4, 95% CI=-2.5, -0.2) and were more likely to have been rated as improved (CGI-I: treatment difference=-0.5, 95% CI=-0.8, -0.1) and as not severely unwell (CGI-S: treatment difference=-0.3, 95% CI=-0.5, 0.0) by the treating clinician. Patients who received CBD also showed greater improvements that fell short of statistical significance in cognitive performance (BACS: treatment difference=1.31, 95% CI=-0.10, 2.72) and in overall functioning (GAF: treatment difference=3.0, 95% CI=-0.4, 6.4). CBD was well tolerated, and rates of adverse events were similar between the CBD and placebo groups. Conclusions: These findings suggest that CBD has beneficial effects in patients with schizophrenia. As CBD's effects do not appear to depend on dopamine receptor antagonism, this agent may represent a new class of treatment for the disorder.
Article
Full-text available
Introduction: The roots of the cannabis plant have a long history of medical use stretching back millennia. However, the therapeutic potential of cannabis roots has been largely ignored in modern times. Discussion: In the first century, Pliny the Elder described in Natural Histories that a decoction of the root in water could be used to relieve stiffness in the joints, gout, and related conditions. By the 17th century, various herbalists were recommending cannabis root to treat inflammation, joint pain, gout, and other conditions. There has been a subsequent paucity of research in this area, with only a few studies examining the composition of cannabis root and its medical potential. Active compounds identified and measured in cannabis roots include triterpenoids, friedelin (12.8 mg/kg) and epifriedelanol (21.3 mg/kg); alkaloids, cannabisativine (2.5 mg/kg) and anhydrocannabisativine (0.3 mg/kg); carvone and dihydrocarvone; N-(p-hydroxy-β-phenylethyl)-p-hydroxy-trans-cinnamamide (1.6 mg/kg); various sterols such as sitosterol (1.5%), campesterol (0.78%), and stigmasterol (0.56%); and other minor compounds, including choline. Of note, cannabis roots are not a significant source of Δ⁹-tetrahydrocannabinol (THC), cannabidiol, or other known phytocannabinoids. Conclusion: The current available data on the pharmacology of cannabis root components provide significant support to the historical and ethnobotanical claims of clinical efficacy. Certainly, this suggests the need for reexamination of whole root preparations on inflammatory and malignant conditions employing modern scientific techniques.
Article
Anecdotal evidence that cannabis preparations have medical benefits together with the discovery of the psychotropic plant cannabinoid Δ9-tetrahydrocannabinol (THC) initiated efforts to develop cannabinoid-based therapeutics. These efforts have been marked by disappointment, especially in relation to the unwanted central effects that result from activation of cannabinoid receptor 1 (CB1), which have limited the therapeutic use of drugs that activate or inactivate this receptor. The discovery of CB2 and of endogenous cannabinoid receptor ligands (endocannabinoids) raised new possibilities for safe targeting of this endocannabinoid system. However, clinical success has been limited, complicated by the discovery of an expanded endocannabinoid system - known as the endocannabinoidome - that includes several mediators that are biochemically related to the endocannabinoids, and their receptors and metabolic enzymes. The approvals of nabiximols, a mixture of THC and the non-psychotropic cannabinoid cannabidiol, for the treatment of spasticity and neuropathic pain in multiple sclerosis, and of purified botanical cannabidiol for the treatment of otherwise untreatable forms of paediatric epilepsy, have brought the therapeutic use of cannabinoids and endocannabinoids in neurological diseases into the limelight. In this Review, we provide an overview of the endocannabinoid system and the endocannabinoidome before discussing their involvement in and clinical relevance to a variety of neurological disorders, including Parkinson disease, Alzheimer disease, Huntington disease, multiple sclerosis, amyotrophic lateral sclerosis, traumatic brain injury, stroke, epilepsy and glioblastoma.
Article
Rapid eye movement (REM) sleep behavior disorder (RBD) is a parasomnia characterized by the presence of a complex of signals resulting from the loss of REM sleep atony and manifested by vigorous and sometimes violent motor jerks and nocturnal vocalizations associated with dream enactment. RBD might be a clinical predictor of severity for Parkinson's disease (PD) and one of its most important non-motor manifestations, preceding the emergence of synucleinopathy by several years or even decades. The detection of RBD may represent a therapeutic window for research regarding the development of new neuroprotective therapies with the potential to modify the natural course of synucleinopathies, such as PD. We performed a scoping review of studies indexed in MEDLINE and LILACS focusing on pharmacological interventions for RBD associated with PD. Fourteen articles were selected. Study designs comprised randomized and non-randomized clinical trials (n = 153 participants) and observational studies (retrospective cohorts and case series, n = 248 participants). Melatonin and clonazepam appear to be useful for treating RBD in PD, but these findings are mostly supported by observational studies and a few controlled studies with a small number of participants. New pharmacological agents, such as melatoninergic agonists and phytocannabinoids, appear to be promising therapies. The findings from studies focusing on anticholinesterases and new dopaminergic agents are still deemed inconclusive.
Article
Background and objective: Falls are a major public health problem in older adults. Earlier studies showed that psychotropic medication use increases the risk of falls. The aim of this study is to update the current knowledge by providing a comprehensive systematic review and meta-analysis on psychotropic medication use and falls in older adults. Methods and design: This study is a systematic review and meta-analysis. A search was conducted in Medline, PsycINFO, and Embase. Key search concepts were "falls," "aged," "medication," and "causality." Studies were included that investigated psychotropics (antipsychotics, antidepressants, anxiolytics, sedatives, and hypnotics) as risk factors for falls in participants ≥60 years of age or participants with a mean age of ≥70 years. Meta-analyses were performed using generic inverse variance method pooling unadjusted and adjusted odds ratio (OR) estimates separately. Results: In total, 248 studies met the inclusion criteria for qualitative synthesis. Meta-analyses using adjusted data showed the following pooled ORs: antipsychotics 1.54 [95% confidence interval (CI) 1.28-1.85], antidepressants 1.57 (95% Cl 1.43-1.74), tricyclic antidepressants 1.41 (95% CI 1.07-1.86), selective serotonin reuptake inhibitors 2.02 (95% CI 1.85-2.20), benzodiazepines 1.42 (95%, CI 1.22-1.65), long-acting benzodiazepines 1.81 (95%, CI 1.05-3.16), and short-acting benzodiazepines 1.27 (95%, CI 1.04-1.56) Most of the meta-analyses resulted in substantial heterogeneity that did not disappear after stratification for population and healthcare setting. Conclusions: Antipsychotics, antidepressants, and benzodiazepines are consistently associated with a higher risk of falls. It is unclear whether specific subgroups such as short-acting benzodiazepines and selective serotonin reuptake inhibitors are safer in terms of fall risk. Prescription bias could not be accounted for. Future studies need to address pharmacologic subgroups as fall risk may differ depending on specific medication properties. Precise and uniform classification of target medication (Anatomical Therapeutic Chemical Classification) is essential for valid comparisons between studies.
Article
Background The Dravet syndrome is a complex childhood epilepsy disorder that is associated with drug-resistant seizures and a high mortality rate. We studied cannabidiol for the treatment of drug-resistant seizures in the Dravet syndrome. Methods In this double-blind, placebo-controlled trial, we randomly assigned 120 children and young adults with the Dravet syndrome and drug-resistant seizures to receive either cannabidiol oral solution at a dose of 20 mg per kilogram of body weight per day or placebo, in addition to standard antiepileptic treatment. The primary end point was the change in convulsive-seizure frequency over a 14-week treatment period, as compared with a 4-week baseline period. Results The median frequency of convulsive seizures per month decreased from 12.4 to 5.9 with cannabidiol, as compared with a decrease from 14.9 to 14.1 with placebo (adjusted median difference between the cannabidiol group and the placebo group in change in seizure frequency, −22.8 percentage points; 95% confidence interval [CI], −41.1 to −5.4; P=0.01). The percentage of patients who had at least a 50% reduction in convulsive-seizure frequency was 43% with cannabidiol and 27% with placebo (odds ratio, 2.00; 95% CI, 0.93 to 4.30; P=0.08). The patient’s overall condition improved by at least one category on the seven-category Caregiver Global Impression of Change scale in 62% of the cannabidiol group as compared with 34% of the placebo group (P=0.02). The frequency of total seizures of all types was significantly reduced with cannabidiol (P=0.03), but there was no significant reduction in nonconvulsive seizures. The percentage of patients who became seizure-free was 5% with cannabidiol and 0% with placebo (P=0.08). Adverse events that occurred more frequently in the cannabidiol group than in the placebo group included diarrhea, vomiting, fatigue, pyrexia, somnolence, and abnormal results on liver-function tests. There were more withdrawals from the trial in the cannabidiol group. Conclusions Among patients with the Dravet syndrome, cannabidiol resulted in a greater reduction in convulsive-seizure frequency than placebo and was associated with higher rates of adverse events. (Funded by GW Pharmaceuticals; ClinicalTrials.gov number, NCT02091375.)