ArticlePDF Available

Psychopathological and Cognitive Effects of Therapeutic Cannabinoids in Multiple Sclerosis: A Double-Blind, Placebo Controlled, Crossover Study

Authors:
  • Italian National Institute for Health, Migration and Poverty (NIHMP)
  • Sapienza University of Rome and IRCCS Neuromed, Italy

Abstract and Figures

To study possible psychopathological symptoms and cognitive deficits, abuse induction, as well as general tolerability and effects on quality of life, fatigue and motor function in cannabis-naïve patients with multiple sclerosis (MS) treated with a free-dose cannabis plant extract (Sativex). In an 8-week, randomized, double-blind, placebo-controlled, parallel group crossover trial, 17 cannabis-naïve patients with MS were assessed at baseline and at the end of the cannabis and placebo phases of the trial (each of 3 weeks) by means of Symptom Checklist-90 Revised, Self-rating Anxiety Scale, Multiple Sclerosis Functional Composite (of which 1 dimension is the Paced Auditory Serial Additional Test that was used to evaluate cognition), Visual Analogue Scale on health-related quality of life, Multiple Sclerosis Impact Scale-29, and Fatigue Severity Scale. Postplacebo versus postcannabinoid scores showed that no significant differences could be detected on all the variables under study. A significant positive correlation was found between Delta-9-tetrahydrocannabinol blood levels and scores at the General Symptomatic Index and at the "interpersonal sensitivity," "aggressive behaviour," and "paranoiac tendencies" subscales of the Symptom Checklist-90 Revised. No serious adverse events, abuse tendencies, or direct withdrawal symptoms were reported. Increased desire for Sativex with secondary depression was reported in 1 subject. Cannabinoid treatment did not induce psychopathology and did not impair cognition in cannabis-naïve patients with MS. However, the positive correlation between blood levels of Delta-9-tetrahydrocannabinol and psychopathological scores suggests that at dosages higher than those used in therapeutic settings, interpersonal sensitivity, aggressiveness, and paranoiac features might arise, although greater statistical power would be necessary to confirm this finding.
Content may be subject to copyright.
Psychopathological and Cognitive Effects of Therapeutic
Cannabinoids in Multiple Sclerosis: A Double-Blind, Placebo
Controlled, Crossover Study
Massimiliano Aragona, MD,* Emanuela Onesti, MD,ÞValentina Tomassini, MD,þ
Antonella Conte, MD,þShiva Gupta, MD,§ Francesca Gilio, MD,þPatrizia Pantano, MD,þ
Carlo Pozzilli, PhD,þand Maurizio Inghilleri, PhD, MDþ
Objectives: To study possible psychopathological symptoms and cog-
nitive deficits, abuse induction, as well as general tolerability and effects
on quality of life, fatigue and motor function in cannabis-naBve patients
with multiple sclerosis (MS) treated with a free-dose cannabis plant
extract (Sativex).
Methods: In an 8-week, randomized, double-blind, placebo-controlled,
parallel group crossover trial, 17 cannabis-naBve patients with MS were
assessed at baseline and at the end of the cannabis and placebo phases of
the trial (each of 3 weeks) by means of Symptom ChecklistY90 Revised,
Self-rating Anxiety Scale, Multiple Sclerosis Functional Composite
(of which 1 dimension is the Paced Auditory Serial Additional Test
that was used to evaluate cognition), Visual Analogue Scale on health-
related quality of life, Multiple Sclerosis Impact Scale-29, and Fatigue
Severity Scale.
Results: Postplacebo versus postcannabinoid scores showed that
no significant differences could be detected on all the variables under
study. A significant positive correlation was found between $-9-
tetrahydrocannabinol blood levels and scores at the General Symp-
tomatic Index and at the Binterpersonal sensitivity,[Baggressive
behaviour,[and Bparanoiac tendencies[subscales of the Symptom
ChecklistY90 Revised. No serious adverse events, abuse tendencies,
or direct withdrawal symptoms were reported. Increased desire for
Sativex with secondary depression was reported in 1 subject.
Conclusions: Cannabinoid treatment did not induce psychopa-
thology and did not impair cognition in cannabis-naBve patients with
MS. However, the positive correlation between blood levels of $-9-
tetrahydrocannabinol and psychopathological scores suggests that at
dosages higher than those used in therapeutic settings, interpersonal
sensitivity, aggressiveness, and paranoiac features might arise, although
greater statistical power would be necessary to confirm this finding.
Key Words: cannabinoids, psychosis, anxiety, cognition, multiple
sclerosis
(Clin Neuropharm 2009;32: 41Y47)
Psychopathological symptoms and cognitive impairment are
often reported as possible side effects of recreational
cannabis use. In population surveys, anxiety and psychotic
symptoms are commonly experienced after cannabis use.
1,2
The
psychotomimetic effect of cannabis, which was proved in ex-
perimental studies reporting transient positive and negative
psychotic symptoms together with other psychopathological
phenomena,
3
is greater in subjects predisposed for psychosis
4,5
and probably in adolescent smokers due to a greater vulnera-
bility of the developing brain.
6
On the other hand, cannabis use
has been frequently reported in anxiety disorders
7
; it might
precipitate psychosis,
5,8
and the overall risk of developing
psychotic symptoms has been described as increasing 3-fold in
cannabis users.
6
The acute administration of cannabis was reported to
produce transient effects on several neuropsychological func-
tions (eg, attention, memory, visual-motor coordination, verbal
fluency, and psychomotor speed).
3,9,10
In Bheavy users[(eg,
subjects smoking several joints per day, during periods of
many years), a long-term, mild cognitive impairment was
reported.
11,12
Although no significant cognitive deficits were
reported in frequent but moderate users of cannabis,
13
the
persistent effects of cannabis on cognition remain uncertain.
14
Cannabinoids have recently been suggested for the treat-
ment of neurodegenerative disorders, vascular and inflamma-
tory diseases, cancer, and central pain,
15
and they are currently
suggested for the treatment of spasticity and chronic pain in
multiple sclerosis (MS). In the light of this potential therapeutic
role, studies focusing on psychological and cognitive tolera-
bility of cannabinoids in MS are eagerly awaited.
16
At the
moment, some studies on general tolerability of cannabis treat-
ment in MS are available,
17
whereas, at least in our knowledge,
no specific data have been published on its cognitive and psy-
chopathological tolerability.
Consequently, the aims of this study were (i) to characterize
the effects of a 3-week cannabinoid treatment with a cannabis
plant extract containing $-9-tetrahydrocannabinol ($-9-THC)
and cannabidiol (CBD) in equal proportions on several psycho-
pathological dimensions, including psychotic symptoms and
anxiety, and cognitive performances in cannabis-naBve patients
with MS; (ii) to evaluate the tolerability and possible abuse
induction; and (iii) to study the effects of cannabis on quality of
lifeYrelated dimensions, on fatigue and motor function.
MATERIALS AND METHODS
Subjects
Seventeen patients with MS were recruited from the MS
Outpatient Clinic at BSapienza[University. After obtaining
written informed consent, subjects underwent a baseline
neurological and psychiatric assessment. To be eligible for
inclusion into this study, patients had to fulfill the following
criteria: male or female subjects between 18 and 60 years of
age; right-handed with normal right-hand function; a baseline
Expanded Disability Status Scale
18
score ranging from 3.5 to
ORIGINAL ARTICLE
Clinical Neuropharmacology &Volume 32, Number 1, January/February 2009 41
*Chair of Philosophy of Psychopathology, BSapienza[University; Division
of Neurology, Neurological Centre of Latium; Department of Neuro-
logical Sciences, BSapienza[University, Rome, Italy; and §Department
of Radiology, Westchester Medical Center, Valhalla, NY.
Address correspondence and reprint requests to Maurizio Inghilleri, PhD,
MD, Dipartimento di Scienze Neurologiche, Viale dell_Universita
`30,
00185 Rome, Italy; E-mail: maurizio.inghilleri@uniroma1.it
Copyright *2009 by Lippincott Williams & Wilkins
DOI: 10.1097/WNF.0b013e3181633497
6.5; a stable disease for at least 30 days before study entry and
nosystemic corticosteroid therapy within 4 weeks of randomi-
zation; significant spasticity in at least 2 muscle groups;
antispastic and immunomodulatory agents (dosage, frequency,
and route of administration) stable, before the study entry, for at
least 1 and 6 months, respectively; no history of epilepsy of
alcohol or substance abuse and no major medical illnesses;
absence of psychiatric disorders or cognitive impairment at first
evaluation; no history of psychiatric disorders; no concomitant
therapy with psychoactive drugs; no female patient who was
pregnant, lactating, or planning pregnancy during the course of
the study; and no previous use of cannabis. This study was
approved by the local Ethical Committee. Patients were in-
formed about the potential for psychosis, anxiety, and panic, and
they gave their informed consent to be included into the trial.
Study Drug
Patients received cannabis-based medicine extract (Sativex,
GW Pharma, UK) presented in a pump action sublingual spray.
Sativex is composed of whole cannabis plant extract, contain-
ing $-9-THC (27 mg/mL) and CBD (25 mg/mL), in ethanol/
propylene glycol (50:50) excipient. Each actuation delivers
100 KL of spray, containing THC 2.7 mg and CBD 2.5 mg.
Placebo had the appearance, smell, and taste of the active
formulation in ethanol/propylene glycol (50:50) excipient but
contained no active components.
Study Design
This 8-week, randomized, double-blind, placebo-controlled,
crossover trial was part of a larger study on functional mag-
netic resonance imaging and electrophysiological correlates
of cannabinoid effect in MS patients experiencing spasticity
(Pozzilli C et al, personal communication).
Patients taking immunomodulatory, antispastic, and/or any
other pharmacological treatment were admitted in the study if
therapy intake was stable for at least 6 and 1 months, respec-
tively, before the study entry and were requested to maintain the
intake of these pharmacological agents at fixed dosage, route,
and time of administration. Trial assessments were scheduled
at fixed times to be performed under standardized identical
conditions (eg, daytime, interval to the last administration of
antispastic, immunomodulatory compounds, and others).
Screening assessment was followed by randomization and
dose introduction. Patients were randomly assigned to 2 counter-
balanced groups starting either with Sativex or with placebo as
the first drug. After 3 weeks, the first treatment was discon-
tinued, and patients entered a washout phase of 2 weeks, before
starting the second treatment phase of 3 weeks. During each
treatment period, all patients had to reach the optimal, individ-
ualized dosage to subjectively relief spasticity. The number of
daily actuations was recorded.
As shown in Figure 1, all patients completed the psycho-
pathological and cognitive measures before starting therapy and
at the end of both Sativex and placebo treatment phases. A
withdrawal was done at the end of each treatment phase to
evaluate the THC and CBD levels in the blood. Patients were
recontacted and asked to estimate their desire for cannabis and
whether they had noted any new medical or psychiatric problem
twice after the end of the study, 2 weeks, and 2 months later,
respectively.
Outcome Measures
At each visit, all patients were asked to complete rating
scales to assess fatigue, disability, psychopathology, cognitive
functioning, and the physical and psychological impact of MS
on quality of life.
Specifically, psychopathological symptoms were assessed
using 2 self-reporting devices: the Symptom ChecklistY90
Revised (SCL-90-R)
19
and the Self-rating Anxiety Scale
(SAS).
20
The SCL-90-R is a 90-item symptom inventory; the
subject rates each item on a 5-point scale of distress, from 0
Bnot-at-all[to 4 Bextremely.[The 90 items are scored and
interpreted in terms of 9 primary symptom dimensions and 1
global severity index of distress named General Symptomatic
Index. The dimensions are labeled as somatization, obsessive-
compulsiveness, interpersonal sensitivity, depression, anxiety,
hostility, phobic anxiety, paranoid ideation, and psychoticism.
The SAS is a 20-item symptom inventory specifically designed
to study anxiety; the subject rates each item on a 4-point scale
of distress, from 1 Bnone or a little of the time[to 4 Bmost or all
of the time.[
Cognitive performances were evaluated using the Paced
Auditory Serial Additional Test (PASAT), 1 of the 3 dimensions
of the Multiple Sclerosis Functional Composite (MSFC).
21
PASAT is a measure of cognitive function that assesses audi-
tory information processing speed and flexibility, as well as
calculation ability. The dependent variable is the sum of cor-
rect answers given by the subject (out of 60 possible). The
other 2dimensions of the MSFC are the Timed 25-Foot Walk
(T25FW), which is a quantitative measure of leg function and
ambulation, and the 9-Hole Peg Test (9HPT), a quantitative
measure of arm and hand function. To calculate the overall
MSFC, scores for each component are converted to zscores
using a specific formula. All these 3 independent clinical
dimensions contribute equally to the overall MSFC score.
Quality of life and physical and psychological impact of
MS from the patients_perspective were studied by means of
3instruments: the Visual Analogue Scale on health-related
quality of life,
22
the Fatigue Severity Scale,
23
and the 29-item
Multiple Sclerosis Impact Scale.
24
Visual Analogue Scale on
health-related quality of life consists of a straight line of a spec-
ified length with verbal descriptors at each end consisting of
short and easily understood phrases that describe the variable
being measured. Fatigue Severity Scale is a 9-item questionnaire
that assesses the effect of fatigue on daily living. Each item is a
FIGURE 1. Study design. 1 = Screening assessment and
randomization; administration of clinical, cognitive, and
psychopathological questionnaires; first-phase treatment
introduction. 2 = First-phase treatment discontinuation;
administration of clinical, cognitive, and psychopathological
questionnaires; first blood withdrawal; assessment of possible side
effects. 3 = Administration of clinical, cognitive, and
psychopathological questionnaires; second-phase treatment
introduction. 4 = Second-phase treatment discontinuation;
administration of clinical, cognitive, and psychopathological
questionnaires; second blood withdrawal; assessment of possible
side effects. 5 = Clinical assessment; evaluation of possible
long-lasting side effects and of possible desire/craving for
cannabinoids. 6 = Clinical assessment; evaluation of possible
long-lasting side-effects and of possible desire/craving for
cannabinoids.
Aragona et al Clinical Neuropharmacology &Volume 32, Number 1, January/February 2009
42 *2009 Lippincott Williams & Wilkins
statement on fatigue that the subject rates from 1 Bcompletely
disagree[to 7 Bcompletely agree.[Patients with a mean score
of 4 or more were defined as suffering from significant fatigue.
The 29-item Multiple Sclerosis Impact Scale is an instrument
measuring the physical (20 items) and psychological (9 items)
impact of MS. The 2 summary scores are generated by sum-
ming individual items and then transformed to a 0 to 100 scale.
High scores indicate worse health.
THC and CBD Plasma Measurement
Tetrahydrocannabinol and CBD levels in the plasma were
measured according to the LC-MS/MS spectrometric method
(Liquid Chromatography with tandem Mass Spectrometer
detection) of Valiveti and Stinchcomb,
25
with slight modifi-
cations. Chromatographic analyses were carried out using a
High-Performance Liquid Chromatography with tandem Mass
Spectrometer detection (HPLC-MS/MS) system. Negative elec-
trospray ionization was used and all analyses were performed
in Multiple Reaction Monitoring (MRM) mode. The transition
were m/z313V245 for THC and CBD. The limit of quantita-
tion was 0.1 ng/mL for $-9-THC and CBD.
Statistical Analysis
We assigned patients to treatment sequence by using an
independent statistician-generated randomization code. Investi-
gators allocated patients consecutively by time of inclusion at
the study site.
9HPT task periods were averaged over the 2 trials for each
hand, averaging together both the dominant and nondominant
hand trials. T25FW trial periods were averaged over the 2 trials.
MSFC scores were calculated using test results from the baseline
visit from all patients. Analyses for MSFC were conducted
without replacing data values as well.
For all variables measured except for MSFC PASAT, 9HPT,
and T25FW, Wilcoxon_s matched pairs signed-rank tests were
used to compare variables measured at the end of each 3-week
treatment period (postplacebo vs post-Sativex) given that all
variables except those listed above are defined by finite scoring
systems that do not fit a normal distribution. For MSFC PASAT,
9HPT, and T25FW, paired ttests were conducted to compare
the variables postplacebo versus post-Sativex levels, given that
these 3 variables are defined by a continuous, normally dis-
tributed time scale.
For correlations with CBD and $-9-THC serum levels, all
dependent variables except for MSFC PASAT, 9HPT, and
T25FW measured during the post-Sativex period were corre-
lated using Spearman rank correlations given the nonparamet-
ric nature of these variables as stated above. For MSFC PASAT,
9HPT, and T25FW values measured during post-Sativex pe-
riods, Pearson correlations were evaluated to correlate with CBD
and $-9-THC serum levels.
All tests are reported with their local Pvalues, thus iden-
tifying differences that would have potential statistical sig-
nificance (PG0.05), if chosen as the primary efficacy criterion.
For the correlation statistics, Bonferroni correction was not
used because the comparisons did not fall within a single joint
family of comparisons by definition.
RESULTS
All the 17 patients who entered the study completed the
trial and were included in the analysis.
Baseline demographic and clinical characteristics are re-
ported in Table 1. Baseline assessment did not reveal significant
cognitive problems. All the included patients had taken
antispastic and immunomodulatory compounds in their clin-
ical history, with generally poor results. During the study,
only 5 patients were taking immunomodulatory compounds
(interferon-beta, copolymer-1, or azatioprine) and 10 were tak-
ing antispastic drugs (baclofen, dantrolene sodium, chlorohyd-
rate tizanidina, or botulinum toxin). All patients maintained the
intake of these pharmacological agents at fixed dosages, route,
and time of administration in relation to the assessment points
of the trial.
The number of daily actuations used was significantly
higher during the placebo than during the Sativex period
(15.16 + 4.51 vs 8.20 + 3.15; t= 5.31, PG0.001).
Postplacebo versus post-Sativex scores are reported in
Table 2. It is shown that neither psychopathological nor
cognitive scores differed between the Sativex and the placebo
period. Although objective measures did not reveal any
cognitive impairment, 11 patients during the Sativex admin-
istration and 2 subjects during the placebo period reported
subjective drowsiness and sense of Bslower-thinking[;1of
the 11 subjects who reported this symptom experienced tran-
sient mental confusion with temporal and spatial disorientation.
Measures of fatigue, leg and arm function, quality of life, and
TABLE 1. Baseline Demographic and Clinical Characteristics
(n = 17)
Characteristics Mean (TSD)
Age (range) 49.8 (T6.64)
Sex (Female/Male) 11:6
Disease duration, yr 20.76 (T8.42)
Disease course 17 SP
EDSS score 6.1 (T0.3)
MSFC (PASAT) 35.76 (T13.22)
MSFC (9HPT) 29.66 (T8.21)
MSFC (T25FW) 26.85 (T14.51)
MSIS-29 (physical items) 53.64 (T11.83)
MSIS-29 (psychological items) 36.54 (T20.51)
FSS 5.61 (T0.97)
VAS QoL 4.29 (T2.08)
SAS 36.38 (T6.42)
SCL-90 GSI 0.63 (T0.28)
SCL-90 Somat 10.61 (T4.17)
SCL-90 Obsess 6.61 (T4.36)
SCL-90 Sens 4.46 (T4.31)
SCL-90 Depr 12.76 (T6.59)
SCL-90 Anx 4.46 (T3.45)
SCL-90 Aggr 2.46 (T1.66)
SCL-90 Phob 4.3 (T3.32)
SCL-90 Para 2.92 (T3.61)
SCL-90 Psychosis 4.23 (T2.42)
SP indicates secondary progressive MS; EDSS, Expanded Disability
Status Scale; MSFC, Multiple Sclerosis Functional Composite; PASAT,
Paced Auditory Serial Additional Test; 9HPT, 9-Hole Peg Test; T25FW,
Timed 25-Foot Walk; MSIS-29, Multiple Sclerosis Impact Scale; FSS,
Fatigue Severity Scale; VAS QoL, Visual Analogue Scale for Quality of
Life; SAS, Self Rating Anxiety Scale; SCL-90, Symptom Checklist-90
Revised; GSI, General Symptomatic Index; Somat, somatized anxiety;
Obsess, obsessive-compulsive features; Sens, sensitivity; Depr, depres-
sion; Anx, anxiety; Aggr, aggressive behaviour; Phob, phobic anxiety;
Para, paranoiac tendencies; Psychosis, psychotic symptoms.
Clinical Neuropharmacology &Volume 32, Number 1, January/February 2009 Effects of Therapeutic Cannabinoids in MS
*2009 Lippincott Williams & Wilkins 43
physical and psychological impairment due to MS did not
differ between the 2 treatment periods.
Correlation between clinical scores and plasma levels of
CBD and $-9-THC are reported in Table 3. A significant
positive correlation was found between the $-9-THC blood
levels and scores at the General Symptomatic Index and at
the BInterpersonal sensitivity,[BAggressive behaviour,[and
BParanoiac tendencies[of the SCL-90-R. No correlations were
found between the other major component of Sativex (CBD)
and the studied variables.
No serious adverse events (hospitalization, death) occurred
during the study. Side effects reported during the placebo
and Sativex phases were generally mild (Table 4). During the
Sativex administration, 1 patient required treatment in the
emergency department because of transient mental confusion
with temporal and spatial disorientation, tachycardia, increased
blood pressure, and mydriasis. Symptoms recovered in few
hours without requiring hospitalization, and the patient was able
to continue the study without further problems.
All patients were recontacted twice after the end of the
study, 2 weeks, and 2 months later, respectively. At the 2-week
but not at the 2-month visit, one 56-year-old lady reported
intense desire to reintroduce Sativex after withdrawal and
developed depressive symptoms (successfully treated with
sertraline 50 mg/die), probably related to the impossibility to
keep taking Sativex. None of the other patients reported
craving for cannabis, dependence, and withdrawal effects, as
well as any new medical or psychiatric problems.
DISCUSSION
The primary aim of this study was to explore the onset
of psychopathological symptoms and cognitive deficits in
cannabis-naBve patients with MS treated with a cannabis plant
extract (Sativex) for relieving their spasticity. General toler-
ability, possible abuse induction, and the effects of canna-
binoids on quality of life, fatigue, and motor function were
also studied.
Our study evaluated the effects of cannabinoids on
cognition by means of PASAT, which is widely used to test
sustained attention, divided attention, concentration, and infor-
mation processing speed in MS patients.
26
We found no
significant differences between Sativex and placebo in the
PASAT scores, although we found a higher frequency of
subjective drowsiness and sense of Bslower-thinking[in the
cannabinoid phase of the study. Tacking into consideration the
TABLE 2. Postplacebo Versus Post-Sativex Clinical Scales Scores (n = 17)
Scales Postplacebo (mean TSD) Post-Sativex (mean TSD) Statistic
MSFC (PASAT) 43.0 (T11.8) 42.4 (T13.6) t= 0.28, P= 0.79
MSFC (9HPT) 28.4 (T8.66) 28.75 (T9.37) t=j0.33, P= 0.75
MSFC (T25FW) 26.2 (T16.9) 25.1 (T15.1) t = 0.51, P= 0.62
MSIS-29 (physical items) 63.0 (T16.8) 62.3 (T13.1) z= 0.57, P= 0.57
MSIS-29 (psychological items) 47.8 (T17.8) 46.3 (T15.9) z=j0.47, P= 0.64
FSS 5.58 (T1.50) 5.89 (T0.93) z=j0.16, P= 0.88
VAS QoL 3.65 (T2.29) 4.00 (T2.00) z= 1.01, P= 0.31
SAS 37.7 (T8.67) 35.7 (T7.69) z=j1.26, P= 0.21
SCL-90 GSI 0.76 (T0.43) 0.77 (T0.43) z=j0.12, P= 0.91
SCL-90 Somat 12.11 (T7.25) 12.94 (T5.62) z= 0.60, P= 0.55
SCL-90 Obsess 7.94 (T6.2) 8.11 (T5.39) z=j0.19, P= 0.85
SCL-90 Sens 4.52 (T4.62) 4.76 (T4.57) z=j0.03, P= 0.98
SCL-90 Depr 16 (T9.13) 15.58 (T8.65) z=j0.45, P= 0.65
SCL-90 Anx 5.41 (T4.78) 6.05 (T5.65) z= 0.65, P= 0.52
SCL-90 Aggr 3.11 (T3.01) 2.29 (T1.89) z=j0.92, P= 0.36
SCL-90 Phob 6.11 (T4.97) 6.64 (T4.85) z= 0.25, P= 0.81
SCL-90 Para 2.88 (T3.46) 3.17 (T3.74) z= 0.36, P= 0.72
SCL-90 Psychosis 5 (T3.8) 4.23 (T3.63) z=j0.46, P= 0.65
Refer to Table 1 for the explanations of abbreviations.
TABLE 3. Relationship Between Plasma Levels of CBME
Components and Clinical Scores
Indexes CBD, ng/mL $-9-THC, ng/mL
MSFC (PASAT) j0.23 (P= 0.37) j0.17 (P= 0.52)
MSFC (9HPT) 0.06 (P= 0.82) j0.01 (P= 0.97)
MSFC (T25FW) 0.07 (P= 0.80) 0.26 (P= 0.33)
MSIS-29 (physical items) 0.14 (P= 0.59) 0.34 (P= 0.19)
MSIS-29 (psychological
items)
0.11 (P= 0.68) 0.17 (P= 0.51)
FSS 0.14 (P= 0.60) 0.24 (P= 0.35)
VAS QoL 0.20 (P= 0.44) j0.01 (P= 0.96)
SAS j0.02 (P= 0.93) 0.19 (P= 0.47)
SCL-90 GSI 0.17 (P= 0.52) 0.52 (P= 0.03)
SCL-90 Somat 0.19 (P= 0.47) 0.46 (P= 0.06)
SCL-90 Obsess 0.23 (P= 0.37) 0.43 (P= 0.09)
SCL-90 Sens 0.42 (P= 0.09) 0.67 (PG0.01)
SCL-90 Depr 0.03 (P= 0.91) 0.36 (P= 0.16)
SCL-90 Anx 0.19 (P= 0.46) 0.44 (P= 0.07)
SCL-90 Aggr 0.30 (P= 0.24) 0.57 (P= 0.02)
SCL-90 Phob j0.19 (P= 0.47) 0.06 (P= 0.81)
SCL-90 Para 0.27 (P= 0.30) 0.59 (P= 0.01)
SCL-90 Psychosis j0.05 (P= 0.86) 0.29 (P= 0.25)
Refer to Table 1 for the explanations of abbreviations.
Aragona et al Clinical Neuropharmacology &Volume 32, Number 1, January/February 2009
44 *2009 Lippincott Williams & Wilkins
high sensitivity of PASAT for revealing cognitive deficits, we
are prone to interpret this finding as of no significant impact
on cognition in our cohort. Although a previous study reported
an improvement on PASAT performances during a period of
2 weeks,
27
our results are in accordance with a 4-week study,
which did not find any difference compared with placebo.
28
Our
findings support the hypothesis that cannabinoids taken as a
treatment for MS symptoms do not affect cognition, at least in
the short term. Note that research on long-term recreational
cannabis users (mean length of regular use 910.3 years) showed
that slower cognitive processing rates in PASAT performances
are more common among long-term rather than short-term
users,
11
suggesting that although cognitively safe, a note of
caution is needed in the long-term administration. Another
aspect to be considered as of potential impact on cognition is
the dosage. Our findings suggest that in the range of thera-
peutic dosages (mean daily puffs 8.20 + 3.15, approximately
corresponding to 22 mg of $-9-THC), cannabinoids do not
significantly affect cognition, and this is in accordance with re-
sults from a study reporting no significant cognitive deficits in
frequent but moderate users of cannabis.
13
Although we did not
find any significant correlation between $-9-THC and CBD
plasma levels and PASAT scores at the end of the Sativex
treatment, which suggests that at these moderate dosages, there
is no dose effect on cognition; cognitive tolerability at higher
dosages cannot be predicted on the basis of our data because the
study was not designed to detect such an effect.
The other major element under study was the effect of
cannabinoid treatment on psychopathology. Psychopathological
symptoms were often found in recreational cannabis users,
1,2
and cannabis use had been frequently reported in anxiety
7
and
psychotic disorders.
29
Our finding that psychopathological
scores did not differ between cannabinoids and placebo ap-
parently contrasts with the report of transient Bpositive[and
Bnegative[psychotic symptoms, perceptual alterations, eupho-
ria, and anxiety in healthy volunteers.
3
However, in that study,
the authors evaluated the acute effect of 2 fixed dosages of
intravenously injected $-9-THC in subjects who had previously
smoked cannabis, whereas in our study patients were cannabis
naBve, they took a free-dosage oral-administered cannabis
extract that also contains CBD, and the effects on psychopa-
thology were evaluated after 3 weeks of treatment. These
differences in the study design make a direct comparison of
these 2 studies difficult. To our knowledge, there was no pre-
vious study specifically focused on the evaluation of psycho-
pathological phenomena in MS patients. However, a few
psychiatric disturbances were reported among other side ef-
fects. In particular, euphoria was the most frequently reported
symptom, ranging from 3.2% to 26.6% of the treated
subjects,
30Y34
whereas other phenomena were found sporad-
ically (hyperactivity,
30
dissociation,
32
feeling Bparanoid,[and
hallucinations
34
).
In our study, euphoria was reported only by 1 patient in the
treatment group (5.8%), and its rate was comparable to that
reported in the placebo group. This finding does not differ
considerably from previous studies. However, neither our nor
previous studies used standardized questionnaires to measure
euphoria, and further studies with more objective and sensitive
measures are thus required.
Having found no differences in the SCL-90 and SAS
scores, our study suggests that in the short term, Sativex ad-
ministered to cannabis-naBve patients for therapeutic purposes
does not increase the risk of main psychopathological symp-
toms. However, we found a moderate but significant positive
correlation between the plasma levels of $-9-THC (the principal
active ingredient of cannabis) and some of the SCL-90 scales.
This suggests that although at the low therapeutic levels used in
this study, cannabinoids do not induce the onset of psycho-
pathological symptoms, at higher dosages, the risk of worsen-
ing interpersonal sensitivity (eg, the tendency of experiencing
feeling of inadequacy in the relationship with others), aggres-
siveness, and paranoiac thoughts cannot be excluded. This
possibility, together with previous reports of sporadic symptoms
like feeling Bparanoid[and hallucinations
34
in patients with MS
treated with cannabinoids, suggests that a careful evaluation of
history of psychosis should be done in all patients addressed
to cannabinoids treatment. Finally, the correlation between
$-9-THC and aggressiveness found in our study contrasts
with a previous research that excluded aggressive feelings in
50 MS patients treated with cannabis-derived drug.
31
Consid-
ering that the relationship between cannabis use and aggres-
siveness is still debated,
35,36
future studies on larger samples
of patients taking cannabinoids as a therapy are needed.
Another aim of the study was the evaluation of the impact
of cannabinoid treatment on some physical problems related to
MS (eg, reduced leg and arm function, fatigue), as well as on
quality of life and physical and psychological impairment. Our
findings suggest that in the short term, none of these variables
changed significantly. These results are in line with current
literature showing that administered for a 2- to 4-week treat-
ment period, cannabis does not significantly improve arms and
legs mobility
27
and fatigue,
37
whereas they differ from those of
Killestein and colleagues,
37
who found a significant worsening
in the total MSFC and 9HPT scores. Our findings also confirm
previous studies showing how cannabis use did not covary with
general scores on quality of life,
37Y39
whereas a positive slight
trend benefit was found on legs motility (T25FW) in long-term
studies.
39
More promising results have been previously reported
with subjective reports: cannabis was reported to relief fatigue
and dysfunction of walking and balance and to improve emo-
tional dysfunction.
40
However, the fact that findings with objec-
tive measurements are mostly conflicting suggests that studies
on larger cohorts are needed to evaluate the impact of canna-
binoid treatment on these variables.
Another issue to be evaluated was the general tolerability.
All subjects included in the study completed the 3 weeks of
trial, and no serious adverse events occurred during the study.
Side effects were slightly more frequent during active treatment
than during placebo. No clinically relevant changes were ob-
served in physical examinations, with the exception of transient
TABLE 4. Side Effects Reported During the 2 Treatment
Periods
Sativex Phase
(n = 17)
Placebo
Phase
(n = 17)
Mouth dryness or burning 5 0
Depression 1 1
Fatigue 6 3
Euphoria 1 1
Headache 3 3
Nausea and vomiting 2 1
Drowsiness and/or slower thinking 11 2
Dizziness and vertigo 8 0
Tremor 1 0
Lower limb weakness 3 0
Clinical Neuropharmacology &Volume 32, Number 1, January/February 2009 Effects of Therapeutic Cannabinoids in MS
*2009 Lippincott Williams & Wilkins 45
increased blood pressure in 1 patient. No changes were found in
hematology or biochemistry parameters. Finally, although some
possible effects of Sativex are similar to those reported with
antispastics (eg, drowsiness, mouth dryness, dizziness, vertigo,
and fatigue) and consequently could have been responsible of
a summatory effect, this was not found in our study.
Taken together, these findings suggest that Sativex was
well tolerated. Long-term studies confirm the good tolerability
profile of cannabinoids for therapeutic use.
17,39
Finally, a crucial point that is usually a matter of concern
when the therapeutic use of cannabinoids is proposed is the
possibility that cannabis may induce dependence and abuse. At
this regard, our study was in a good position to observe these
possible phenomena, being on cannabis-naBve subjects and
allowing for free dosages. Interestingly, none of the patients
abused the use of Sativex during the treatment phase, the mean
daily dosage being quite low (23Y25 mg) and the number of
daily actuations being significantly higher in the placebo than
in the cannabis phase (suggesting that when patients had
reached the therapeutic daily dosage, they spontaneously inter-
rupted the dosage augmentation of Sativex without craving
induction, whereas in the case of placebo, they continued to
increase the number of daily actuations trying to reach active
effects). Different motivations of the user (recreational vs
therapeutic) might partially explain our findings that cannabi-
noid treatment in MS does not induce tolerability and dosage
increase. However, in the case of other psychoactive sub-
stances (eg, benzodiazepines, tramadol, and zolpidem), some
tendency to induce tolerability is also found in therapeutic
settings with consequent increased risk of dosage augmenta-
tion and dependence,
41Y44
suggesting that our findings may be
not com pletely due to the above-mentioned factor. A related
but different factor is that in recreational use of cannabis, the
route of administration (inhalation) usually maximizes the
Cmax and shortens the Tmax in order to increase the psy-
choactive effect, whereas Sativex (sublingual/oromucosal
administration) reaches a low Cmax and has a fairly long
Tmax.
45
Different pharmacokinetics might be responsible for
less gratifying subjective experiences due to the delayed
psychoactive effect of Sativex, thus explaining (at least in
part) why patients were not driven to increase the dosage.
Furthermore, direct withdrawal symptoms were not
reported, as expected.
17
However, some days after the end of
the trial, 1 patient experienced desire to reintroduce Sativex.
Whether this desire was driven by a perceived effective treat-
ment (she explained it as a reaction to the impossibility to
continue the treatment that she considered very helpful, and in
effect, she presented a significant leg mobility improvement,
with a 57% drop of T25FW scores) or whether it represented
a withdrawal symptom cannot be addressed by a single case.
Two months after the termination of the study, all the subjects
included in the study had not increased desire to be treated
again with Sativex and did not present any new medical or
psychiatric problem. However, long-term studies are needed in
larger samples to evaluate the possible dependence induced
by cannabinoid therapy.
There are some limitations in our study, which need to
be considered in the interpretation of our findings. The sample
size was quite small to address completely all the questions
arising from a study evaluating the psychopathological impact
of cannabinoids. However, the power of the study was though
to evaluate the effect of Sativex on physical symptoms (eg,
spasticity) and its electrophysiological and imaging counter-
part. The study duration of treatment was relatively short,
again aimed to provide pilot information about the study drug.
As a consequence, direct generalization of these data to larger
clinical populations is not viable, and more focused studies are
needed to investigate the safety of cannabinoids on cognitive
and psychopathological domains in patients with MS. Cog-
nitive performances were studied with 1 test (PASAT), and
no other information about cognition in this cohort was avail-
able. More detailed test batteries should be included in future
studies. Finally, the oromucosal route of administration of
cannabis might have induced unpredictable interindividual and
intraindividual variability in plasma $-9-THC levels, not
detectable with blood tests.
In conclusion, cannabinoids prescribed for therapeutic
reasons to cannabis-naBve patients with MS did not induce
onset of psychotic or anxiety symptoms and did not impair
cognition. Safety and tolerability were generally good, drug
tolerance and dose increasing were not reported during the
trial, and desire for Sativex or abuse was not present at follow-
up. However, the positive correlation between blood levels
of $-9-THC and psychopathological scores suggests that at
dosages higher than those used in therapeutic settings, inter-
personal sensitivity, aggressiveness, and paranoiac features
might arise. Finally, the effects of cannabinoids on quality of
life, fatigue, and motor function of MS patients were non-
significant, suggesting that the effective impact of cannabi-
noids in these patients as well as the most proper target
symptoms remain to be better ascertained.
ACKNOWLEDGMENTS
Sativex was kindly provided by GW Pharma Ltd, Salisbury,
England. The grant has been supported as the Project of
University Research (ex-quota 60%)Vyear 2004Vby the
University BSapienza[of Rome.
REFERENCES
1. Thomas H. A community survey of adverse effects of cannabis use.
Drug Alcohol Depend 1996;42:201Y207.
2. Stefanis NC, Delespaul P, Henquet C, et al. Early adolescent cannabis
exposure and positive and negative dimensions of psychosis. Addiction
2004;99:1333Y1341.
3. D_Souza DC, Perry E, MacDougall L, et al. The psychotomimetic effect
of intravenous delta-9-tetrahydrocannabinol in healthy individuals:
implications for psychosis. Neuropsychopharmacology 2004;29:
1558Y1572.
4. Henquet C, Krabbendam L, Spauwen J. Prospective cohort study of
cannabis use, predisposition for psychosis, and psychotic symptoms in
young people. BMJ 2005;330:11.
5. Degenhardt L, Hall W. Is cannabis use a contributory cause of
psychosis? Can J Psychiatry 2006;51:556Y565.
6. Semple DM, McIntosh AM, Lawrie SM. Cannabis as a risk factor for
psychosis: a systematic review. J Psychopharmacol 2005;19:187Y194.
7. Dannon PN, Lowengrub K, Amiaz R, et al. Comorbid cannabis use
and panic disorder: short term and long term follow-up study. Hum
Psychopharmacol 2004;19:97Y101.
8. Iversen L. Long-term effects of exposure to cannabis. Curr Opin
Pharmacol 2005;5:69Y72.
9. Curran HV, Brignell C, Fletcher S, et al. Cognitive and subjective
dose-response effects of acute oral Delta 9-tetrahydrocannabinol
(THC) in infrequent cannabis users. Psychoparmacol (Berl)
2002;164:61Y70.
10. O_Leary DS, Block RI, Turner BM, et al. Marijuana alters the human
cerebellar clock. Neuroreport 2003;14:1145Y1151.
11. Solowij N, Stephens RS, Roffman RA, et al. Cognitive functioning of
Aragona et al Clinical Neuropharmacology &Volume 32, Number 1, January/February 2009
46 *2009 Lippincott Williams & Wilkins
long-term heavy cannabis users seeking treatment. JAMA 2002;287:
1123Y1131.
12. Messinis L, Kyprianidou A, Malefaki S, et al. Neuropsychological
deficits in long-term frequent cannabis users. Neurology 2006;66:
737Y739.
13. Jager G, Kahn RS, Van Den Brink W, et al. Long-term effects of
frequent cannabis use on working memory and attention: an fMRI
study. Psychopharmacol (Berl) 2006;185:358Y368.
14. Verdejo-Garcia A, Lopez-Torrecillas F, Gimenez CO, et al. Clinical
implications and methodological challenges in the study of the
neuropsychological correlates of cannabis, stimulant, and opioid
abuse. Neuropsychol Rev 2004;14:1Y41.
15. Williamson EM, Evans FJ. Cannabinoids in clinical practice. Drugs
2000;60:1303Y1314.
16. Killestein J, Uitdehaag BM. Cannabinoids in multiple sclerosis:
urgent need for long term trials. J Neurol Neurosurg Psychiatry
2005;76:1612.
17. Wade DT, Makela PM, House H, et al. Long-term use of a
cannabis-based medicine in the treatment of spasticity and other
symptoms in multiple sclerosis. Mult Scler 2006;12:639Y645.
18. Kurtzke JF. Rating neurologic impairment in multiple sclerosis: an
expanded disability status scale (EDSS). Neurology
1983;33:1444Y1452.
19. Derogatis LR. The SCL-90-R: Administration Scoring and Procedures
Manual II. Baltimore: Clinical Psychometric Research; 1992.
20. Zung WWK. A rating instrument for anxiety disorders. Psychosomatics
1971;12:371.
21. Fischer JS, Rudick RA, Cutter GR, et al. The Multiple Sclerosis
Functional Composite Measure (MSFC): an integrated approach to MS
clinical outcome assessment. National MS Society Clinical Outcomes
Assessment Task Force. Mult Scler 1999;5:244Y250.
22. Parkin D, Rice N, Jacoby A, et al. Use of a visual analogue scale in a
daily patient diary: modelling cross-sectional time-series data on
health-related quality of life. Soc Sci Med 2004;59:351Y360.
23. Herlofson K, Larsen JP. Measuring fatigue in patients with Parkinson_s
diseaseVthe Fatigue Severity Scale. Eur J Neurol 2002;9:595Y600.
24. Hobart J, Lamping D, Fitzpatrick R, et al. The Multiple Sclerosis Impact
Scale (MSIS-29): a new patient-based outcome measure. Brain
2001;124:962Y973.
25. Valiveti S, Stinchcomb AL. Liquid chromatographic-mass spectrometric
quantitation of Delta9-tetrahydrocannabinol and two metabolites in
pharmacokinetic study plasma samples. J Chromatogr B Analyt Technol
Biomed Life Sci 2004;803:243Y248.
26. Rosti E, Hamalainen P, Koivisto K, et al. The PASAT performance
among patients with multiple sclerosis: analyses of responding patterns
using different scoring methods. Mult Scler 2006;12:586Y593.
27. Vaney C, Heinzel-Gutenbrunner M, Jobin P, et al. Eff icacy, safety and
tolerability of an orally administered cannabis extract in the treatment
of spasticity in patients with multiple sclerosis: a randomized,
double-blind, placebo-controlled, crossover study. Mult Scler 2004;
10:417Y424.
28. Rog DJ, Nurmikko TJ, Friede T, et al. Randomized, controlled trial
of cannabis-based medicine in central pain in multiple sclerosis.
Neurology 2005;65:812Y819.
29. Green B, Young R, Kavanagh D. Cannabis use and misuse prevalence
among people with psychosis. Br J Psychiatry 2005;187:306Y313.
30. Svendsen KB, Jensen TS, Bach FW. Does the cannabinoid dronabinol
reduce central pain in multiple sclerosis? Randomised double blind
placebo controlled crossover trial. BMJ 2004;31:253.
31. Vaney C, Heinzel-Gutenbrunner M, Jobin P, et al. Efficacy, safety and
tolerability of an orally administered cannabis extract in the treatment
of spasticity in patients with multiple sclerosis: a randomized,
double-blind, placebo-controlled, crossover study. Mult Scler
2004;10:417Y424.
32. Rog DJ, Nurmikko TJ, Friede T, et al. Randomized, controlled trial of
cannabis-based medicine in central pain in multiple sclerosis.
Neurology 2005;65:812Y819.
33. Collin C, Davies P, Mutiboko IK, et al. Sativex Spasticity in MS
Study Group. Randomized controlled trial of cannabis-based medicine
in spasticity caused by multiple sclerosis. Eur J Neurol 2007;14:
290Y296.
34. Chong MS, Wolff K, Wise K, et al. Cannabis use in patients with
multiple sclerosis. Mult Scler 2006;12:646Y651.
35. Monshouwer K, Van Dorsselaer S, Verdurmen J, et al. Cannabis use
and mental health in secondary school children. Findings from a
Dutch survey. Br J Psychiatry 2006;188:148Y153.
36. Arendt M, Rosenberg R, Fjordback L, et al. Testing the self-medication
hypothesis of depression and aggression in cannabis-dependent
subjects. Psychol Med 2007;37:935Y945.
37. Killestein J, Hoogervorst EL, Reif M, et al. Safety, tolerability, and
efficacy of orally administered cannabinoids in MS. Neurology
2002;58:1404Y1407.
38. Ventegodt S, Merrick J. Psychoactive drugs and quality of life. Sci
Wo rl d J 2003;18:694Y706.
39. Zajicek JP, Sanders HP, Wright DE, et al. Cannabinoids in multiple
sclerosis (CAMS) study: safety and efficacy data for 12 months
follow up. J Neurol Neurosurg Psychiatry 2005;76:1664Y1669.
40. Consroe P, Musty R, Rein J, et al. The perceived effects of smoked
cannabis on patients with multiple sclerosis. Eur Neurol 1997;3:44Y48.
41. Khong E, Sim MG, Hulse G. Benzodiazepine dependence. Aust Fam
Phys 2004;33:923Y926.
42. Simon GE, Ludman EJ. Outcome of new benzodiazepine prescriptions
to older adults in primary care. Gen Hosp Psychiatry 2006;28:374Y378.
43. Brinker A, Bonnel RA, Beitz J. Abuse, dependence, or withdrawal
associated with tramadol. Am J Psychiatry 2002;159:881.
44. Aragona M. Abuse, dependence, and epileptic seizures after zolpidem
withdrawal: review and case report. Clin Neuropharmacol 2000;23:
281Y283.
45. Guy GW, Robson PJ. A phase 1, open-label, four way crossover
study to compare the pharmacokinetic profiles of a single dose of
29mg of a cannabis-based medicine extract (CBME) administered on
3 different areas of the buccal mucosa and to investigate the
pharmacokinetics of CBME per oral in healthy male and female
volunteers. J Cannabis Ther 2003;3/4:121Y152.
Clinical Neuropharmacology &Volume 32, Number 1, January/February 2009 Effects of Therapeutic Cannabinoids in MS
*2009 Lippincott Williams & Wilkins 47
... Russo et al. [82] conducted a small, open-label trial in 20 patients with MS divided equally between groups with and without neuropathic pain. Subjects were permitted to use Sativex ® (average dose 8 sprays/day) for 4 weeks. ...
... Antidepressants, e.g., amitriptyline and duloxetine, and anticonvulsants, e.g., gabapentin and pregabalin, are effective in treating neuropathic pain in some patients, but this condition remains an enormous area of unmet clinical need. Sativex ® has undergone clinical testing as a potential treatment for neuropathic pain characterized by allodynia [85], diabetic neuropathy [86], and neuropathic pain associated with MS [77,81,82]. In addition, Sativex ® has also been evaluated for the management of pain caused by rheumatoid arthritis [87]. ...
... In addition, Sativex ® has also been evaluated for the management of pain caused by rheumatoid arthritis [87]. The positive effects of Sativex ® in treating neuropathic pain caused by MS [77,81,82] were discussed in the previous section. Nurmikko et al. [85] evaluated the efficacy and safety of Sativex ® in a 1:1 randomized, double-blind, placebo-controlled 5-week clinical trial in 125 subjects with neuropathic pain of peripheral origin. ...
Article
Full-text available
The potential of botanical cannabinoids as potential anti-inflammatory and neuroprotective agents.
... In previous research, Sativex has not been demonstrated to have any negative impact on cognition or mood, and it did not impair driving abilities either [45,178]. Individual sensitivity, aggression, and paranoid traits may appear at dosages higher than those utilized in therapeutic settings, according to Aragona et al. [179], who observed a positive link between THC blood levels and aberrant results on psychological examinations. After a month of Sativex treatment, Russo et al. [177] observed psychotic symptom onset (including suicidal ideation) in one patient who was taking a relatively high daily drug dosage (12 vs. 9 puffs), had a baseline ZSS (a 20-item self-report assessment device built to measure anxiety levels) score of 47, and was being treated with other psychoactive drugs [180]. ...
Article
Sativex is a cannabis-based medicine that comes in the form of an oromucosal spray. It contains equal amounts of Δ9-tetrahydrocannabinol and cannabidiol, two compounds derived from cannabis plants. Sativex has been shown to have positive effects on symptoms of amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS), and sleep disorders. It also has analgesic, antiinflammatory, antitumoral, and neuroprotective properties, which make it a potential treatment option for other neurological disorders. The article reviews the results of recent preclinical and clinical studies that support the therapeutic potential of Sativex and the molecular mechanisms behind its neuroprotective benefits in various neurological disorders. The article also discusses the possible advantages and disadvantages of using Sativex as a neurotherapeutic agent, such as its safety, efficacy, availability, and legal status.
... We found 14 reports eligible for our review, including the registered trial protocols for 5 of the studies [29][30][31][32][33]. Nine articles (1510 patients) were included in the systematic review and 7 (1135 patients) provided data for meta-analysis [6,[34][35][36][37][38][39]. The selection process is detailed in Fig. (1). ...
Article
Background: Spasticity affects 54% of multiple sclerosis (MS) patients at disease onset, but this rate gradually increases with disease progression. Spasticity does not fully respond to standard treatment in one-third of the patients. Objective: Our systematic review and meta-analysis assessed whether add-on nabiximols, can improve MS-associated refractory spasticity. Methods: The systematic literature search was performed in Web of Science, MEDLINE, Scopus, CEN- TRAL, and Embase, on 15/10/2021, without restrictions. We included in the review blinded, randomized, placebo-controlled trials evaluating the efficacy of nabiximols in adult MS patients with refractory spas- ticity, by comparison with placebo. The primary outcome was responder rate by spasticity numerical rat- ing scale (NRS). Secondary outcomes were spasticity-related parameters. We used random effect models to calculate odds ratios (OR) or mean differences and the corresponding 95% CI. Bias-factors were as- sessed with Cochrane risk of bias tool (RoB2). (PROSPERO ID: CRD42021282177). Results: We identified 9 eligible articles, of which 7 (1128 patients) were included in the meta-analysis. The spasticity numerical rating scale (NRS) was significantly higher in the nabiximols group than in the placebo group (OR 2.41 (95% CI 1.39; 4.18)). Secondary outcomes were in accordance with our primary results. At least some concerns were detected in the risk of bias analysis. Conclusion: Our results indicate that nabiximols is efficient in MS associated spasticity, refractory to standard treatment and it may be considered as add-on symptomatic therapy. Nevertheless, further studies are needed to establish the optimal treatment protocol - dose, duration, moment of initiation, disease type.
Article
Cannabis use and cannabis use disorders have taken on a new social significance as a result of partial legalization. In 2021 a total of 4.5 million adults (8.8%) in Germany used the drug. The number of users as well as problematic use have risen in the last decade. Cannabis products with a high delta-9-tetrahydrocannabinol (THC) content and their regular use lead to changes in cannabinoid receptor distribution in the brain and to modifications in the structure and functionality of relevant neuronal networks. The consequences of cannabinoid use are particularly in the psychological functioning and can include intoxication, harmful use, dependence with withdrawal symptoms and cannabis-induced mental disorders. Changes in the diagnostics between ICD-10 and ICD-11 are presented. Interdisciplinary S3 guidelines on cannabis-related disorders are currently being developed and will be finalized shortly.
Article
Objetivo: Analisar sobre os efeitos dos canabinoides no tratamento e/ou alívio de sintomas de doenças neurodegenerativas, como: Parkinson, Doença de Alzheimer e Esclerose Múltipla. Métodos: Foi realizada uma revisão integrativa intencionando compilar os principais resultados do uso dos canabinoides na prevenção e controle de doenças neurodegenerativas com foco nas doenças de Parkinson, Alzheimer e Esclerose Múltipla. Foram selecionados artigos científicos disponíveis no portal Periódicos Capes distribuídos nas bases de dados PubMed, Science Direct e Medline. Resultados: Foram encontrados 331 artigos científicos filtrados apenas para testes clínicos, destes após triagem foram lidos integralmente 52 textos e para esta revisão foram selecionados 21 artigos. Os artigos selecionados demonstram resultados positivos na melhora de sintomas e na qualidade de vida geral em todas as patologias. A maioria dos estudos foram realizados em países europeus, demonstrando a necessidade de maiores investimentos em pesquisas clínicas com canabinoides principalmente na América do Sul. Considerações finais: Com a análise dos textos é possível concluir que os canabinoides Tetra-hidrocanabinol (THC) e Canabidiol (CBD) são compostos benéficos em testes clínicos com resultados positivos no controle e prevenção de sintomas das três doenças neurodegenerativas escolhidas para esta revisão.
Article
Objetivo: Analisar sobre os efeitos dos canabinoides no tratamento e/ou alívio de sintomas de doenças neurodegenerativas, como: Parkinson, Doença de Alzheimer e Esclerose Múltipla. Métodos: Foi realizada uma revisão integrativa intencionando compilar os principais resultados do uso dos canabinoides na prevenção e controle de doenças neurodegenerativas com foco nas doenças de Parkinson, Alzheimer e Esclerose Múltipla. Foram selecionados artigos científicos disponíveis no portal Periódicos Capes distribuídos nas bases de dados PubMed, Science Direct e Medline. Resultados: Foram encontrados 331 artigos científicos filtrados apenas para testes clínicos, destes após triagem foram lidos integralmente 52 textos e para esta revisão foram selecionados 21 artigos. Os artigos selecionados demonstram resultados positivos na melhora de sintomas e na qualidade de vida geral em todas as patologias. A maioria dos estudos foram realizados em países europeus, demonstrando a necessidade de maiores investimentos em pesquisas clínicas com canabinoides principalmente na América do Sul. Considerações finais: Com a análise dos textos é possível concluir que os canabinoides Tetra-hidrocanabinol (THC) e Canabidiol (CBD) são compostos benéficos em testes clínicos com resultados positivos no controle e prevenção de sintomas das três doenças neurodegenerativas escolhidas para esta revisão.
Article
Objective: Nabiximols is used for treating various symptoms associated with multiple sclerosis (MS). Nabiximols is also being investigated as a potential treatment medication for individuals with cannabis use disorder (CUD). A variety of investigations have shown that, at low doses, nabiximols is overall well tolerated for MS treatment. However, due to tolerance, the management of CUD would likely require much higher doses of nabiximols to be effective. The effects of high doses of nabiximols on clinical laboratory tests remain unclear. Therefore, we investigated the sub-chronic effects of high doses of nabiximols on liver function, renal function, and other routine blood tests in this prospective study. Methods: We performed a secondary analysis of various blood markers results collected during a double-blind, placebo-controlled randomized clinical trial (Sativex and Behavioral-relapse Prevention Strategy in Cannabis Dependence, NCT01747850, https://clinicaltrials.gov/ct2/show/record/NCT01747850). This trial tested the impact of the 12-week administration of nabiximols with a maximum daily dose of up to 113.4 mg THC/105 mg CBD. Results: The measurements of the various biomarkers were in the normal range during the 12-week time frame. The results indicate an overall good tolerability of high-dose nabiximols on the blood markers measured. Conclusion: Our preliminary results suggest that high doses of nabiximols might be well tolerated by individuals with CUD.
Chapter
Multiple sclerosis (MS) is a chronic demyelinating disease of the central and peripheral nervous systems. The symptoms, such as vision problems, muscle weakness, pain, balance problems, and paralysis, are due to the uncontrolled or inappropriate neural transmission that gradually worsens as the disease progresses. During the early stage of the disease, patients may experience long symptom-free periods, and the periods are interrupted by flare-ups that last days to weeks. It has been indicated that Cannabis, delta-9-tetrahydrocannabinol (THC), nabiximols, and oral Cannabis extract (OCE) may diminish spasticity associated with MS [19]. Nabiximols is a mucosal spray containing delta-9-THC and cannabidiol (CBD) in the 1:1 ratio and has been developed for treating MS. The US FDA has approved nabiximols and has also been approved in several European countries, Canada, and New Zealand for treating spasticity associated with MS.
Article
Full-text available
Purpose of Review The use of cannabinoid-based medicines in symptomatic treatment for people with multiple sclerosis (MS) is high. This review aimed to assess benefit and harms of cannabinoids, including synthetic, herbal and plant-derived cannabinoids, for reducing symptoms in adults with MS. Recent Findings Cannabinoids exert effects of potential relevance to MS: their action of neuroinflammation suppression, their immunomodulatory and neuroprotective effects in the central nervous system could benefit people with MS. Nabiximols is approved and available for people with MS and moderate or severe spasticity in many countries, and cannabinoids are also licensed in several countries for the treatment of chronic neuropathic pain. Summary Compared with placebo, cannabinoids likely reduce the severity of spasticity and improve well-being in the short term. Their effect on chronic neurological pain is uncertain. Serious adverse events are not increased; however, withdrawals due to adverse events are slightly greater in the cannabinoid group than in the placebo group. Most of the included trials had major weaknesses. Studies of high quality, with large sample sizes and longer follow-up period, are needed. Moreover, further research is needed in order to assess the short-and long-term adverse effects of these drugs.
Article
Full-text available
This Phase I, open label, four-way crossover study pertains to pharmacokinetic parameters of four cannabis based medicine extracts (CBME). Sublingual, buccal and oro-pharyngeal test treatments (GW-1000-02) consisted of 25 mg cannabidiol (CBD) + 25 mg Δ-tetra-hydrocannabinol (THC) per ml formulated in ethanol (eth):propylene glycol (PG) (50:50), with peppermint flavouring with a 100 μl actuation volume (total dose 10 mg CBD + 10 mg THC in 4 actuations). An oral capsule contained 2.5 mg CBD + 2.5 mg THC sprayed onto granulated lactose and encapsulated in soft gelatin capsules (total dose of 10 mg CBD + 10 mg THC 4 capsules). This study was performed in healthy volunteers in an open label, 4 period, 3-way randomised crossover followed by a nonrandomised oral dose using single doses of 20 mg of CBME (10 mg CBD + 10 mg THC). In Periods 1 to 3, the test treatment was administered as a liquid spray according to the randomisation scheme (i.e., sublingually, buccally, oro-pharyngeally). In Period 4 the test treatment was delivered as an oral capsule. There was a six-day washout between each dose.Primary objectives were to compare the pharmacokinetic profiles of cannabis based medicine extract (CBME) when administered on different areas of the buccal mucosa. Secondary objectives were to investigate the pharmacokinetic profile of CBME when administered as an oral capsule.Concentrations of THC were higher than the corresponding levels of CBD at most time points. Concentrations of 11-hydroxy-THC exceeded the corresponding concentration of THC at most time points. By 720 min (12 h) post-dose, mean concentrations of each cannabinoid were still above the lower limit of quantification (LLOQ). There was a high degree of inter-subject and intra-subject variability in the plasma concentrations achieved.Tmax of CBD and THC occurred earlier following sublingual administration than oro-pharyngeal or buccal although only the difference in Tmax of CBD compared with buccal was statistically significant. Cmax of both CBD and THC was greatest following buccal administration although this was not statistically significant. AUC was greatest following oro-pharyngeal and was statistically significantly greater than buccal. The lower bioavailability, as measured by AUC, following buccal administration when compared to the sublingual and oro-pharyngeal routes may be related to the difficulty of spraying onto the inside of the cheek reported during the study and could be due to some loss of spray. Buccal administration of the pump action sublingual spray (PASS) test treatment resulted in a later Tmax but greater Cmax when compared to the sublingual and oro-pharyngeal routes. Comparison of the sublingual and oro-pharyngeal routes showed no statistically significant difference in THC or CBD pharmacokinetic parameters other than an earlier Tmax following sublingual dosing. The oral capsule appeared to show an early Tmax of both CBD and THC. Mean Cmax of THC and 11-hydroxy-THC were greater, but in contrast the Cmax of CBD was lower, than following the PASS treatments. Relative to THC, the plasma level AUC of 11-hydroxy-THC was proportionally greatest following oral capsules which could be a reflection of greater metabolism by this route. Of the PASS treatments the ratio of 11-hydroxy-THC to THC was greatest following sublingual and least following oro-pharyngeal. There was very wide inter-and to a lesser extent intra-subject variability in pharmacokinetics. Differences in mean values between the routes of administration, even when statistically significant, are small relative to the very wide range of values between subjects. The sublingual and oro-pharyngeal routes of administration appear to have the same pharmacokinetic results. The buccal pharmacokinetic parameters are lower when compared to the sublingual and oro-pharyngeal routes.A total of 146 adverse events (AEs) occurred in 12 subjects. Two events were classified as moderate (flu-like illness and pharyngeal irritation) and the remaining 144 were classified as mild. All routes of administration were well tolerated by all subjects with no serious AEs and no withdrawals due to AEs.The overall results indicate that administration of the liquid spray (GW-1000-02) need not be limited to sublingual administration. The oral capsule, has good bioavailability, and provided, as is the case here the formulation is not oil based, may be a viable formulation when self-titration is not necessary.
Article
Various lines of evidence suggest an association between cannabis and psychosis. Five years ago, the only significant case-control study addressing this question was the Swedish Conscript Cohort. Within the last few years, other studies have emerged, allowing the evidence for cannabis as a risk factor to be more systematically reviewed and assessed. Using specific search criteria on Embase, PsychINFO and Medline, all studies examining cannabis as an independent risk factor for schizophrenia, psychosis or psychotic symptoms, published between January 1966 and January 2004, were examined. Additional studies were also reviewed from references found in retrieved articles, reviews, and a cited reference search (ISI-Web of Science). Studies selected for meta-analysis included: (i) case-control studies where exposure to cannabis preceded the onset of schizophrenia or schizophrenia-like psychosis and (ii) cohort studies of healthy individuals recruited before the median age of illness onset, with cannabis exposure determined prospectively and blind to eventual diagnosis. Studies of psychotic symptoms were also tabulated for further discussion. Eleven studies were identified examining the relationship between cannabis use and psychosis. Seven were included in the meta-analysis, with a derived odds ratio (fixed effects) of 2·9 (95% confidence interval = 2.4-3.6). No evidence of publication bias or heterogeneity was found. Early use of cannabis did appear to increase the risk of psychosis. For psychotic symptoms, a dose-related effect of cannabis use was seen, with vulnerable groups including individuals who used cannabis during adolescence, those who had previously experienced psychotic symptoms, and those at high genetic risk of developing schizophrenia. In conclusion, the available evidence supports the hypothesis that cannabis is an independent risk factor, both for psychosis and the development of psychotic symptoms. Addressing cannabis use, particularly in vulnerable populations, is likely to have beneficial effects on psychiatric morbidity.
Article
Changes in health policy have underlined the importance of evidence-based clinical practice and rigorous evaluation of patient-based outcomes. As patient-based outcome measurement is particularly important in treatment trials of multiple sclerosis, a number of disease-specific instruments have been developed recently. One limitation of these instruments is that none was developed using the standard psychometric approach of reducing a large item pool generated from people with multiple sclerosis. Consequently, an outcome measure for clinical trials of multiple sclerosis that is disease specific and combines patient perspective with rigorous psychometric methods will complement existing instruments. The aim of this study was to develop such a measure. Standard psychometric methods were used. A pool of 129 questionnaire items was generated from interviews with 30 people with multiple sclerosis, expert opinion and literature review. The questionnaire was administered by postal survey to 1530 people selected randomly from the Multiple Sclerosis Society membership database. Redundant items and those with limited measurement properties were removed. The remaining items (n = 41) were grouped into scales using factor analysis, and then refined to form the Multiple Sclerosis Impact Scale (MSIS-29), an instrument measuring the physical (20 items) and psychological (nine items) impact of multiple sclerosis. Five psychometric properties of the MSIS-29 (data quality, scaling assumptions, acceptability, reliability and validity) were examined in a separate postal survey of 1250 Multiple Sclerosis Society members. A preliminary responsiveness study of the MSIS-29 was undertaken in 55 people admitted for rehabilitation and intravenous steroid treatment of relapses. The MSIS-29 satisfied all psychometric criteria. Data quality was excellent, missing data were low (maximum 3.9%), item test–re-test reliability was high (r = 0.65–0.90) and scale scores could be generated for >98% of respondents. Item descriptive statistics, item convergent and discriminant validity, and factor analysis indicated that it was legitimate to generate scores for MSIS-29 scales by summing items. MSIS-29 scales showed good variability, small floor and ceiling effects, high internal consistency (Cronbach's alpha ≤0.91) and high test–re-test reliability (intraclass correlation ≤0.87). Correlations with other measures and the analysis of group differences provided evidence that the MSIS-29 measures the physical and psychological impact of multiple sclerosis. Effect sizes (physical scale = 0.82, psychological scale = 0.66) demonstrated preliminary evidence of good responsiveness. These results indicate the MSIS-29 is a clinically useful and scientifically sound patient-based outcome measure of the impact of multiple sclerosis suitable for clinical trials and epidemiological studies.
Article
Context Cognitive impairments are associated with long-term cannabis use, but the parameters of use that contribute to impairments and the nature and endurance of cognitive dysfunction remain uncertain.Objective To examine the effects of duration of cannabis use on specific areas of cognitive functioning among users seeking treatment for cannabis dependence.Design, Setting, and Participants Multisite retrospective cross-sectional neuropsychological study conducted in the United States (Seattle, Wash; Farmington, Conn; and Miami, Fla) between 1997 and 2000 among 102 near-daily cannabis users (51 long-term users: mean, 23.9 years of use; 51 shorter-term users: mean, 10.2 years of use) compared with 33 nonuser controls.Main Outcome Measures Measures from 9 standard neuropsychological tests that assessed attention, memory, and executive functioning, and were administered prior to entry to a treatment program and following a median 17-hour abstinence.Results Long-term cannabis users performed significantly less well than shorter-term users and controls on tests of memory and attention. On the Rey Auditory Verbal Learning Test, long-term users recalled significantly fewer words than either shorter-term users (P = .001) or controls (P = .005); there was no difference between shorter-term users and controls. Long-term users showed impaired learning (P = .007), retention (P = .003), and retrieval (P = .002) compared with controls. Both user groups performed poorly on a time estimation task (P<.001 vs controls). Performance measures often correlated significantly with the duration of cannabis use, being worse with increasing years of use, but were unrelated to withdrawal symptoms and persisted after controlling for recent cannabis use and other drug use.Conclusions These results confirm that long-term heavy cannabis users show impairments in memory and attention that endure beyond the period of intoxication and worsen with increasing years of regular cannabis use.
Article
The imidazopyridine zolpidem is a short-acting hypnotic chemically distinct from benzodiazepines (BZs). According to its peculiar neuropharmacologic activity (selectivity for the omega 1-BZ receptors), zolpidem is expected to be a pure hypnotic, without the other effects of BZs. In particular, it has been stressed that zolpidem is well tolerated in adults and in the elderly, and that tolerance, abuse, dependence, rebound insomnia, and other withdrawal effects do not develop in relation to zolpidem administration. However, despite these assumptions, zolpidem abuse, dependence, and withdrawal effects have been recently discussed and reviewed herein. In addition, the case of a 43-year-old woman who had an epileptic attack after abrupt interruption of an abused, high dose of zolpidem (600 mg/d), is reported and discussed. At the clinical level, it is stressed that the subjective effects of zolpidem are comparable to those of other BZs, and that abuse, dependence, and withdrawal seizures cannot be avoided simply shifting the regimen of a BZ abuser to zolpidem. At the pharmacologic level, it is important to note that zolpidem's clinical effects cannot be explained on the basis of the old distinction between omega 1 and 2 receptors because this distinction is no longer valid; the new classification of GABA(A) receptor subtypes is reported and zolpidem activity at this level is discussed herein.
Article
Primary objectives of this study were to assess the pharmacokinetic characteristics of CBME when administered sublingually in different ratios, to determine if the pharmacokinetic profiles of THC and its metabolite 11-hydroxy-THC are different when administered sublingually in different formulations, and to characterise the pharmacokinetic profile of CBD when administered with THC in equal amounts. Secondary objectives were to determine if there was a correlation between intoxication levels and plasma concentrations of THC and/or its metabolite 11-hydroxy-THC, and to assess safety and tolerability of CBME when administered sublingually.Methodology employed a double-blind, randomised, three-way crossover study of placebo, High THC and CBD:THC administered sublingually as a liquid spray. Twenty-four subjects were planned, dosed, completed the study and were analysed.Test products were Δ-tetrahydrocannabinol (THC, formulated as 25 mg THC per ml) with or without cannabidiol (CBD) (formulated as 25 mg CBD + 25 mg THC per ml) formulated in ethanol (Eth):propylene glycol (PG) with peppermint (ppmt) flavouring or matching placebo, administered with a 100 μl pump. Each subject received one single dose of 10 mg THC and one single dose of 10 mg CBD + 10 mg THC plus a single dose of placebo in a randomised manner on three separate occasions. The washout period was six days between each dose. Placebo was Eth:PG in a 50:50 ratio with ppmt flavouring, administered with a 100 μl actuator pump.Mean plasma concentrations show that following administration of both High THC and CBD:THC formulations CBD and or THC was detectable in plasma in measurable concentrations 15–30 minutes after dosing, although individual subjects showed quite wide variability, 15 to 135 minutes, to appearance measurable concentrations. At all time points up to 180 minutes after dosing mean concentrations of THC were greater following the High THC formulation than CBD:THC. Concentrations of THC were also greater than corresponding concentrations of CBD following the CBD:THC treatment.There were no statistically significant differences in mean Cmax, t1/2, AUC0-t and AUC0-∞of both THC and 11-hydroxy-THC between the High THC and CBD:THC formulations. THC Tmax was statistically significantly later following CBD:THC than High THC (p = 0.014) and this was the only statistically significant difference in pharmacokinetic parameters between the treatments. The AUC values (AUC0-t and AUC0-∞) for THC show an approximate 8 to 10-fold difference between the lowest and highest subject values while the difference for CBD was approximately 3.5 to 4-fold. Differences in Cmax were 20 to 30 fold for THC and approximately 14-fold for CBD. Intra-subject differences in values for THC between treatments were smaller though differences in Cmax of up to 5-fold and 3-fold in AUC (AUC0-t and AUC0-∞) were observed. Other than a single isolated significant difference in Tmax there were no significant differences in pharmacokinetic parameters between the CBD:THC and High THC formulations. The bioavailability of THC appears to be greater than that of CBD.Mean intoxication scores on both CBME treatments were very low throughout the observation period. The majority of subjects scored zero for the majority of assessment points and there were few scores greater than three on the Box Scale 11 (BS-11). Recorded intoxication scores do not seem to show a direct relationship to plasma concentrations of THC and/or 11-hydroxy-THC either within or between subjects. The time of intoxication scores in individual subjects do not seem to relate consistently with the timing of increases in plasma concentrations or maximal concentrations of THC or 11-hydroxy-THC. Neither is there an apparent relationship between subjects reporting intoxication and those with the highest plasma levels of THC or 11-hydroxy-THC.No subjects withdrew from the study as a result of adverse events and both active and the placebo test treatments were well tolerated. The treatment with the least number of treatment related adverse events was placebo. High THC and CBD:THC had a greater number of subjects who experienced intoxication type adverse events and application site type reactions. The most common overall adverse event experienced was throat irritation, followed by dizziness, somnolence, oral paraesthesia and then headache. All the events were mild and only two events needed any treatment. There were no clinically significant changes from baseline for haematology, biochemistry, vital signs or ECGs.There was wide inter- and intra-subject variability in pharmacokinetic parameters with up to 10-fold differences in THC AUC between subjects and even greater differences in Cmax. Results suggest that there are no overall statistically significant differences between the pharmacokinetic parameters of High THC and CBD:THC other than a delay in Tmax. Considering the wide inter- and intra-subject variability in pharmacokinetic parameters including Tmax this is unlikely to be clinically important in a medication that is self titrated by the patient.