ArticlePDF Available

Sativex® as Add-on therapy Vs. further optimized first-line ANTispastics (SAVANT) in resistant multiple sclerosis spasticity: a double-blind, placebo-controlled randomised clinical trial

Authors:

Abstract and Figures

Purpose/aim: To evaluate the efficacy of tetrahydrocannabinol [THC]:cannabidiol [CBD] oromucosal spray (Sativex®) as add-on therapy to optimized standard antispasticity treatment in patients with moderate to severe multiple sclerosis (MS) spasticity. Methods: Sativex as Add-on therapy Vs. further optimized first-line ANTispastics (SAVANT) was a two-phase trial. In Phase A, eligible patients received add-on THC:CBD spray for 4 weeks to identify initial responders (≥ 20% improvement from baseline in spasticity 0-10 numerical rating scale [NRS] score). Following washout, eligible initial responders were randomised to receive THC:CBD spray or placebo for 12 weeks (double-blinded, Phase B). Optimization of underlying antispasticity medications was permitted in both groups across all study periods. Results: Of 191 patients who entered Phase A, 106 were randomised in Phase B to receive add-on THC:CBD spray (n = 53) or placebo (n = 53). The proportion of clinically-relevant responders after 12 weeks (≥ 30% NRS improvement; primary efficacy endpoint) was significantly greater with THC:CBD spray than placebo (77.4 vs 32.1%; P < 0.0001). Compared with placebo, THC:CBD spray also significantly improved key secondary endpoints: changes in mean spasticity NRS (P < 0.0001), mean pain NRS (P = 0.0013), and mean modified Ashworth’s scale (P = 0.0007) scores from Phase B baseline to week 12. Adverse events, when present, were mild/moderate and without new safety concerns. Conclusions: Add-on THC:CBD oromucosal spray provided better and clinically relevant improvement of resistant MS spasticity compared with adjusting first-line antispasticity medication alone.
Content may be subject to copyright.
Full Terms & Conditions of access and use can be found at
http://www.tandfonline.com/action/journalInformation?journalCode=ines20
International Journal of Neuroscience
ISSN: 0020-7454 (Print) 1543-5245 (Online) Journal homepage: http://www.tandfonline.com/loi/ines20
Sativex® as Add-on therapy Vs. further optimized
first-line ANTispastics (SAVANT) in resistant
multiple sclerosis spasticity: a double-blind,
placebo-controlled randomised clinical trial
Jolana Markovà, Ute Essner, Bülent Akmaz, Marcella Marinelli, Christiane
Trompke, Arnd Lentschat & Carlos Vila Silván
To cite this article: Jolana Markovà, Ute Essner, Bülent Akmaz, Marcella Marinelli, Christiane
Trompke, Arnd Lentschat & Carlos Vila Silván (2018): Sativex® as Add-on therapy Vs. further
optimized first-line ANTispastics (SAVANT) in resistant multiple sclerosis spasticity: a double-
blind, placebo-controlled randomised clinical trial, International Journal of Neuroscience, DOI:
10.1080/00207454.2018.1481066
To link to this article: https://doi.org/10.1080/00207454.2018.1481066
Accepted author version posted online: 24
May 2018.
Submit your article to this journal
Article views: 1
View related articles
View Crossmark data
Publisher: Taylor & Francis
Journal: International Journal of Neuroscience
DOI: https://doi.org/10.1080/00207454.2018.1481066
Sativex® as Add-on therapy Vs. further optimized first-line
ANTispastics (SAVANT) in resistant multiple sclerosis
spasticity: a double-blind, placebo-controlled randomised
clinical trial
Jolana Markovàa, Ute Essnerb, Bülent Akmazc, Marcella Marinellid, Christiane Trompkec, Arnd
Lentschatc, Carlos Vila Silváne
a Neurology Department, Thomayer's Hospital, Praha, Czech Republic
b O. Meany Consultancy GmbH, Hamburg, Ohkamp 26, Germany
c International Clinical Trial Managers, Almirall Hermal GmbH, Scholtzstraβe 3, Reinbeck, Germany
d Clinical Statistics, Almirall S.A., Laureà Miró, 408 08980 Sant Feliu de Llobregat, Barcelona, Spain
e Neurology Medical Manager, Global Medical Affairs, Almirall S.A., Barcelona, Spain
Correspondence:
Jolana Markovà
Thomayer's Hospital
Vídeňská 800
Praha 4 - 140 59 Czech Republic
jolana.markova@ftn.cz
Word count (body): ~3870 to end of Discussion
Figures: 4; Tables: 4; References: 22
Language: British English
Keywords: Multiple sclerosis; spasticity; Sativex; THC; CBD; nabiximols
Abstract
Purpose/aim: To evaluate the efficacy of tetrahydrocannabinol [THC]:cannabidiol [CBD] oromucosal
spray (Sativex®) as add-on therapy to optimized standard antispasticity treatment in patients with
moderate to severe multiple sclerosis (MS) spasticity.
Methods: Sativex as Add-on therapy Vs. further optimized first-line ANTispastics (SAVANT) was a
two-phase trial. In Phase A, eligible patients received add-on THC:CBD spray for 4 weeks to identify
initial responders (≥ 20% improvement from baseline in spasticity 0-10 numerical rating scale [NRS]
score). Following washout, eligible initial responders were randomised to receive THC:CBD spray or
placebo for 12 weeks (double-blinded, Phase B). Optimization of underlying antispasticity
medications was permitted in both groups across all study periods.
Results: Of 191 patients who entered Phase A, 106 were randomised in Phase B to receive add-on
THC:CBD spray (n = 53) or placebo (n = 53). The proportion of clinically-relevant responders after 12
weeks (≥ 30% NRS improvement; primary efficacy endpoint) was significantly greater with THC:CBD
spray than placebo (77.4 vs 32.1%; P < 0.0001). Compared with placebo, THC:CBD spray also
significantly improved key secondary endpoints: changes in mean spasticity NRS (P < 0.0001), mean
pain NRS (P = 0.0013), and mean modified Ashworth’s scale (P = 0.0007) scores from Phase B
baseline to week 12. Adverse events, when present, were mild/moderate and without new safety
concerns.
Conclusions: Add-on THC:CBD oromucosal spray provided better and clinically relevant
improvement of resistant MS spasticity compared with adjusting first-line antispasticity medication
alone.
Introduction
Spasticity is a common chronic symptom in patients with multiple sclerosis (MS) which increases in
prevalence and severity as the disease progresses [13]. It is frequently accompanied by pain,
spasms, mobility restrictions, sleep disturbances, and/or bladder dysfunction and is strongly
associated with fatigue, anxiety, and depression [4,5]. Patients quality of life worsens as spasticity
severity increases [68].
Interventional procedures (e.g. physiotherapy) and pharmacological therapy are the main
approaches for treating MS spasticity. Baclofen and tizanidine are recommended first-line
pharmacological options in the European Union (EU) [4,9], and dantrolene is indicated in certain
countries (albeit used less often). However, treatment with these conventional oral antispasticity
medications is often limited by undesired adverse effects, including effects on the central nervous
system (CNS), increased risk of falls, and/or by a waning effect as MS progresses [10]. About one-
third of MS patients continue to experience moderate to severe spasticity despite first-line
treatment [1113], and a relevant proportion of patients and physicians are dissatisfied with
standard antispasticity medications [7].
Randomised clinical trials [1416] and observational studies conducted in routine clinical practice
[17,18] have shown that an oromucosal spray of tetrahydrocannabinol (THC) and cannabidiol (CBD)
(Sativex®, Nabiximols USAN name) is an effective and well-tolerated option for treating resistant MS
spasticity. THC:CBD spray is approved in several countries (e.g. Canada, Germany, Italy, Spain, UK)
and across Europe it is indicated as add-on therapy for patients with moderate to severe MS
spasticity who have not responded adequately to first-line antispasticity medications [19]. This
indication has raised a clinical question in terms of how much greater the benefits of THC:CBD spray
might be compared with those achieved by attempting to further optimize standard first-line oral
antispasticity therapy.
Herein, we report results from the Sativex as Add-on therapy Vs. further optimized first-line
ANTispastics (SAVANT) randomised, placebo-controlled trial which was designed to evaluate the
therapeutic efficacy of add-on THC:CBD spray compared with further optimization of standard
antispasticity therapy in patients with moderate to severe MS spasticity who were not achieving
adequate symptomatic relief after use of two or more optimized first-line antispasticity medications.
Methods
Design and participants
SAVANT was a prospective, randomised, parallel group, double-blind, placebo-controlled two-phase
trial. Patients were enrolled at 15 sites, 14 in the Czech Republic and 1 in Austria.
Main inclusion criteria were: adults ≥ 18 years of age, with a diagnosis of MS and existing MS
spasticity symptoms for at least 12 months; moderate to severe MS spasticity defined as a score of
4 on the MS spasticity 0-10 numerical rating scale (NRS) scale; previous treatment with at least two
different optimized oral MS spasticity therapies which included oral baclofen and/or oral tizanidine
(as monotherapy or in combination therapy); currently receiving optimized treatment with one or
more oral antispasticity drugs (baclofen and/or tizanidine and/or dantrolene as monotherapy or in
combination therapy) for at least 3 months prior to screening without adequate relief of MS
spasticity symptoms. Optimization was defined as reporting achievement of the most effective and
best tolerated dose possible according to approved labelling. Patients provided written informed
consent to participate.
Exclusion criteria included prior administration of THC:CBD spray; current consumption of cannabis
herb or other cannabinoid-based drugs within 30 days prior to study entry; treatment with
botulinum toxin injection for spasticity relief within the previous 6 months; medical history or family
history of major psychiatric disorders other than depression; known or suspected history of a
dependence disorder or heavy alcohol consumption; possibility of pregnancy or lactation; history of
myocardial infarction or clinically significant cardiac dysfunction; clinically significant impaired renal
function or impaired hepatic function.
The study design is illustrated in Figure 1. In a single-blind, 4-week, trial period (Phase A), patients
received THC:CBD spray as add-on therapy to optimized standard antispasticity medication. Patients
up-titrated the dosage of THC:CBD spray to a maximum of 12 sprays/day according to posology in
the approved label [19] until optimized symptom relief was achieved. Initial responders were
identified based on having achieved a Minimal Clinically Important Difference (MCID) in MS
spasticity, defined as ≥ 20% improvement from baseline in the MS spasticity 0-10 NRS score, which
was rounded up from the 18% improvement calculated as a MCID [20]. Non-responders were
removed from the study. Initial responders at 4 weeks entered a 1 to 4 week washout phase
designed to minimize carry-over effects, during which THC:CBD spray was withdrawn but underlying
standard antispasticity treatment was continued. Initial responders whose improvement in the MS
spasticity NRS score during Phase A was reduced by ≥ 80% during the washout period were eligible
for Phase B. In Phase B, patients were randomised in a double-blind manner to treatment with
THC:CBD spray or placebo for 12 weeks. Patients were advised to re-up-titrate their study
medication to the optimal individual dose identified in Phase A, then to maintain the study
treatment at this dose while allowing for adjustments according to the patient’s needs. Optimization
of underlying antispasticity medications was permitted across all study periods.
The study comprised seven clinic visits: screening (Visit 1); start of single-blind THC:CBD spray trial
period in Phase A (Baseline visit, Visit 2); start of washout phase (Visit 3); start of randomised
double-blind treatment in Phase B (Visit 4); then at 4-weekly intervals during the 12-week treatment
period (Visits 5 until 7, end of treatment). Patients therefore participated in the study for a
maximum total duration of 18 to 22 weeks (Figure 1).
The study was conducted in accordance with the recommendations set out in the Declaration of
Helsinki (Seoul 2008) and in compliance with the ICH Consolidated Guideline for Good Clinical
Practice and applicable local laws and regulations.
The EudraCT allocated number was 2015-004451-40.
Outcomes
The primary efficacy endpoint was the proportion of responders after 12 weeks of randomised
treatment in Phase B, where responder was defined as a patient who achieved 30% improvement
(i.e. a clinically important difference [CID]) in the MS spasticity 0-10 NRS score from Phase B baseline
[20].
Secondary efficacy variables were measures of spasticity and associated symptoms during the 12-
week randomised treatment period (Phase B). Changes referred to values at study end compared
with values at Phase B baseline:
 Change from baseline in MS spasticity 0-10 NRS score
 Change from baseline in pain 0-10 NRS score
 Change from baseline in modified Ashworth scale (MAS) score
 Change from baseline in Expanded Disability Status Scale (EDSS) score.
Other secondary efficacy endpoints were: frequency and severity of spasms; sleep disruption 0-10
NRS score; modified Ashworth scale score per muscular group; Barthel activities of daily living (ADL)
index; short form 36 quality of life (QoL) health survey (SF-36); global assessment of clinical change
(GIC) by subject (SGIC) and physician (PGIC); and timed 10-metre walk test. Safety and tolerability
outcomes were collected during the Phase A, washout, and Phase B periods and assessed separately.
The number of patients with adverse events (AEs) and serious adverse events (SAEs); the numbers of
AEs, SAEs, and treatment interruptions related to AEs; and discontinuation rates and the reasons for
discontinuation were also recorded. AEs were assessed for their intensity (mild, moderate, severe)
and causal relationship with study treatment.
AEs were coded using the Medical Dictionary for Regulatory Activities (MedDRA; version 19.1) and
tabulated by system organ class (SOC) and preferred term (PT).
Statistical analysis
A sample size of 82 randomised (1:1 ratio) evaluable patients in Phase B was estimated to provide
90% power to detect a difference of 35% between arms in the primary endpoint. A blinded interim
analysis was performed to ensure adequacy of the sample size calculation.
Analysis of the primary endpoint was performed for the Phase B intention-to-treat (ITT) population.
An analysis was performed using the Phase B per protocol (PP) population to assess the robustness
of the trial. All other efficacy variables were analysed using both the ITT and PP populations. Safety
outcomes were analysed for the safety population.
The Phase B primary efficacy variable was analysed using a logistic regression model, with
baseline value as a covariate and treatment group as factor. Missing data were handled
using Last Observation Carried Forward (LOCF) and Generalized Linear Mixed Models
(GLMM) methods for binary repeated data with GLIMMIX SAS procedure as a sensitivity
analysis.
All secondary continuous efficacy variables with repeated measures were analysed by
means of a mixed model for repeated measures (MMRM). The dependent variable was the
change from baseline to each scheduled post-baseline visit (encompassing all available
measurements in a patient) during the treatment period. The model adjusted for baseline
value as covariate and treatment, visit and treatment-by-visit interaction as fixed effect
factors.
Treatment effects and treatment comparisons were estimated by Least Square (LS) means
and differences in LS means on the treatment-by-visit interaction at the corresponding
visits, along with standard errors (SE) and 95% confidence intervals (CIs), and the p-value
corresponding to the between-treatment group difference.
Finally, secondary binary efficacy variables with repeated measures were analysed using a
GLMM model for binary repeated data with GLIMMIX SAS procedure. All secondary
variables or other outcomes with one post-baseline assessment were analysed using an
observed cases approach.
Continuous data were summarised using descriptive statistics and categorical data are
presented using frequency (n) and percentage (%). All statistical hypotheses were tested at
the two-sided 5% significance level (α=0.05), and corresponding 95% CIs are reported as
appropriate.
Other secondary endpoints, exploratory endpoints and safety endpoints are presented using
descriptive statistics.
Statistical analyses were performed using SAS statistical analysis software Version 9.1.3 or higher
(SAS Institute Inc., Cary, NC, USA).
Results
The demographic and clinical characteristics of patients entering the Phase A trial period (n = 191)
are summarised in Table 1. The population was predominantly female (70.2%) with a mean (SD) age
of 51.3 ± 10.2 years. Most patients had secondary progressive MS (n = 92; 48.2%) or relapsing
remitting MS (n = 78; 40.8%). At baseline, patients had significant disability (mean EDSS score 5.9)
and moderate to severe MS spasticity (mean NRS score 6.4). Patients had a long history of MS and of
MS spasticity with a mean duration of 14.2 and 7.8 years, respectively.
Most patients in the Phase A safety population had received baclofen (99.0%) and/or tizanidine
(89.0%) as prior antispasticity medication, and 77.5% had taken a combination of baclofen and
tizanidine. No patient had received dantrolene as prior antispasticity medication. Other previous
antispasticity medications taken by patients included other centrally acting agents (14.6%) and
benzodiazepine derivatives (8.4%). At Phase A baseline, 82.2% of patients were receiving baclofen
and 34.5% were receiving tizanidine (inclusive of patients receiving combination therapy). The
demographic and clinical characteristics of Phase B patients were similar to those in the Phase A
population. At Phase B baseline, 84.9% of patients were receiving baclofen, 31.1% were receiving
tizanidine, and 16.0% were receiving combination therapy.
Patient disposition during all phases of the study is shown in Figure 2. Following trial therapy with
THC:CBD spray in Phase A, 134 patients (70.5%) were initial responders ( 20% NRS improvement).
Of these, 106 patients (i.e. 55.5% of 191 Phase A patients) had a ≥ 80% reduction of their Phase A
NRS improvement during the washout period and were eligible for randomization into Phase B. A
total of 50/53 patients (94.3%) allocated to THC:CBD spray and 46/53 patients (88.8%) allocated to
placebo completed 12 weeks of double-blind treatment.
Efficacy
The primary efficacy endpoint, the proportion of MS spasticity 0-10 NRS CID responders after 12
weeks of randomised treatment, was significantly higher in the THC:CBD oromucosal spray group
(41/53; 77.4%) than in the placebo group (17/53; 32.1%), with an adjusted Odds Ratio of 7.0 (95% CI:
2.95 16.74; P < 0.0001; ITT population; Figure 3).
At week 4 in Phase B, 81.1% of patients allocated to THC:CBD spray had reached the initial response
threshold of ≥ 20% NRS improvement versus 45.3% in the placebo group (P = 0.0007).
The mean (SD) number of sprays/day of THC:CBD spray was 7.7 (3.0) at week 4 of Phase A (n =188)
and 7.5 (2.6) at week 4 of randomised Phase B (n = 102). At the week 12 final visit (study end), the
mean (SD) number of sprays/day was 7.3 (2.7) for THC:CBD spray (n = 48) and 8.5 (3.0) for placebo (n
= 46).
Mean (SD) doses of underlying antispasticity study medication were 35.8 (25.5) mg for baclofen and
5.2 (3.0) for tizanidine in Phase A and 35.4 (25.5) mg for baclofen and 5.1 mg (2.9) for tizanidine in
Phase B. During Phase B, seven patients (two in the THC:CBD oromucosal spray group and five in the
placebo group) re-adjusted their baclofen doses.
The results of secondary efficacy outcomes are shown in Table 2. At week 12 in Phase B, significant
reductions were observed with THC:CBD spray versus placebo in the mean MS spasticity 0-10 NRS
score (P < 0.0001; Figure 4), mean pain 0-10 NRS score (P < 0.0013), and mean 0-4 MAS score (P <
0.0007). The difference between THC:CBD spray and placebo in the change of mean MS EDSS scores
from Phase B baseline to week 12 was not significant. Among other secondary efficacy assessments,
THC:CBD spray was significantly superior to placebo for spasms severity (P = 0.0001), sleep
disruption (P = 0.0006), and MAS scores for seven of 10 tested muscular groups (elbow flexor,
extensor and pronator, hip adductor, knee flexor and extensor and foot plantar).
Statistically significant improvements in SGIC and PGIC were reported for THC:CBD vs. placebo after
4 weeks, but tended to decrease at follow up visits (Table 2). Most other secondary efficacy
endpoints changes were in favour of THC:CBD oromucosal spray but comparisons did not reach
statistical significance.
Safety
Features of treatment emergent adverse events (TEAEs) occurring in the Phase A and washout
period safety population (n = 191) and Phase B safety population (n = 106) are shown in Tables 3 and
4, respectively.
In Phase A, 75 AEs (28 moderate, 47 mild) were reported in 46 patients (24.1%), of which 64 events
in 37 patients (19.4%) were considered to be related to study medication (Table 3). The three SAEs
which occurred in two patients (erysipelas; olecranon bursitis and MS relapse) were considered
unrelated to study treatment. There were 54 AE-related treatment interruptions in 29 patients
(15.2%) and five AEs in four patients (2.1%) which led to withdrawal.
The most frequently reported TEAEs during Phase A were: vertigo (15 TEAEs in 14 patients [7.3%]),
somnolence (six TEAEs in three patients [1.6%]), dizziness (four TEAEs in four patients [2.1%]),
diarrhoea (four TEAEs in four patients [2.1%]) and nausea (four TEAEs in four patients [2.1%]). All
TEAEs except one event of vertigo were assessed as related to study treatment.
In the washout phase, 20 TEAEs in 12 patients were ongoing (i.e. carried over from Phase A or
started during washout phase), of which 12 TEAEs in six patients (carried over from Phase A) were
assessed as related to study medication. All TEAEs ongoing during washout were mild or moderate in
intensity. The most frequently reported TEAEs were classified in the SOC Nervous system disorders
(four TEAEs in four patients). All TEAEs were reported by single patients, except for dry mouth, dry
throat and hypertension, which were reported by two patients.
During the 12-week randomised treatment phase (Phase B), no major or new safety concerns were
identified for THC:CBD spray (Table 4).
There was no statistically significant difference between treatment groups in the number of patients
with TEAEs (including SAEs) during Phase B: 19 TEAEs in 12 patients in the active group and 8 TEAEs
in 7 patients in the placebo group. All TEAEs were of either mild or moderate intensity except for
one severe SAE reported in the placebo group.
There was no statistically significant difference between treatment groups in the number of patients
with SAEs during Phase B: one SAE (haematuria of moderate severity) in one patient in the active
group and one SAE (tubulointerstitial nephritis of severe intensity) in one patient in the placebo
group, both of which began and ended in Phase B and were assessed as not related to study
medication.
There was no statistically significant difference between treatment groups in the number of patients
with TEAEs related to study medication during Phase B: seven TEAEs in five patients in the active
group and one TEAE in one patient in the placebo group. All related TEAEs in the active group were
of mild intensity, except for one event of vertigo which was of moderate intensity. Of TEAEs deemed
related to THC:CBD spray, five TEAEs in three patients were classified in the SOC Nervous system
disorders (PT: somnolence, hypoaesthesia, hypogeusia and psychomotor skills impaired). There were
three treatment interruptions related to AEs in two patients (3.8%) in the active group and none in
the placebo group.
Discussion
In patients with moderate-to-severe resistant MS spasticity who initially responded to THC:CBD
spray during a 4-week trial period, adding THC:CBD spray to already-optimized antispasticity
treatment is a better alternative to readjusting the first-line antispasticity medication alone. After 12
weeks treatment, a significantly greater proportion of patients treated with add-on THC:CBD spray
than placebo achieved clinically relevant improvement (≥ 30% NRS improvement) in MS spasticity,
with the difference representing a +45.3% therapeutic gain in favour of THC:CBD oromucosal spray.
Compared with placebo, THC:CBD spray also produced significantly greater reductions in key
secondary efficacy measures including MS spasticity 0-10 NRS, pain 0-10 NRS, and MAS scores. Other
secondary efficacy measures significantly in favour of THC:CBD spray included spasms severity, sleep
disruption, and MAS for seven of 10 tested muscular groups. Although improvement was observed
in the physical activity subscale of the SF-36 in the group treated with THC:CBD spray, overall QoL
scores did not differ significantly between treatment groups, probably due to underlying MS and the
presence of other MS-related symptoms (e.g. fatigue).
In accordance with standard procedures for pre-approval regulatory studies in which changes in
factors other than target medication must be kept to a minimum, no alterations to underlying
antispasticity medications were permitted in previous clinical development trials of THC:CBD spray
[15,16,21]. However, daily clinical experience with THC:CBD spray indicates that patients can adapt
their underlying antispasticity medications according to individual needs and, indeed, continuous
optimization is viewed as an integral component of gaining maximum benefit from a given
treatment. The current study therefore set out to evaluate the efficacy of add-on THC:CBD spray
under conditions closer to daily clinical practice by explicitly allowing optimization of patients’
underlying antispasticity therapy throughout all phases of the study and also ensuring that all
participants had tried and failed at least two first-line antispasticity drugs.
The demographic and clinical characteristics of the patient population were comparable to those in
previous clinical trials and observational studies of THC:CBD spray in resistant MS spasticity [1618],
allowing meaningful comparisons to be made between the studies.
At the end of the 4-week trial period of THC:CBD spray, 70.5% of patients (134/190) were initial
responders (≥ 20% NRS improvement). This was higher than the initial response rate of 47%
(272/572 patients) reported in the enriched-design phase 3 clinical trial of THC:CBD spray [16] but
identical to the initial response rate of 70.5% reported in the Italian health authorities prospective e-
registry study of THC:CBD spray (n = 1615) [18]. The washout period eliminated 28 initial responders
(20.9%) who failed to show ≥ 80% reduction in their Phase A NRS improvement. The inclusion of a
washout period, and requirement for patients’ spasticity levels to return to near pre-treatment
levels, is the main point of difference between our study and that of Novotna et al. [16] and ensured
that treatment effects observed in Phase B were not confounded by any ‘carryover’ or other effects
of THC:CBD spray from Phase A.
After 12 weeks of randomised treatment in initial responders, the CID responder rate was
significantly in favour of THC:CBD spray over placebo (77.4 vs 32.1%; p<0.0001), demonstrating a
broad therapeutic gain despite having allowed for dosage adjustments of underlying antispasticity
medication in both groups. This result compares favourably with respective values of 74% and 51%
(OR: 2.73; 95% CI 1.59 to 4.69; P = 0.0003) reported in the pivotal trial of Novotna et al. (2011)
where the absence of a washout period between Phases A and B may have contributed to a more
pronounced placebo effect [16].
Among study participants who were initial responders to THC:CBD spray in Phase A and met
eligibility criteria after washout for randomization in Phase B, 77.4% of those allocated to THC:CBD
spray in Phase B achieved the CID threshold of ≥ 30% NRS improvement [20]. Thus, the probability of
becoming a CID responder to THC:CBD spray upon starting treatment (start of trial period) was 43%,
which is similar to the 36% reported in the phase 3 trial [16] and to the 41% reported in the Mobility
ImproVEment (MOVE) 2 observational study in Germany [17].
Secondary effectiveness measures in favour of THC:CBD spray, specifically changes from Phase B
baseline to study end in MS spasticity 0-10 NRS, pain 0-10 NRS, and MAS scores, further support the
difference observed between THC:CBD spray and placebo in the responder rate at 12 weeks
(primary endpoint) and are consistent with or superior to results reported for these endpoints in the
previous enriched design trial and observational studies of THC:CBD spray conducted under
approved conditions [16,17]. Although no changes were observed with THC:CBD spray or placebo on
the MS EDSS, this is not surprising given that the EDSS is linked to underlying overall MS evolution, is
largely ambulation driven, and has been shown to have low sensitivity to alterations in MS spasticity
severity [22]. Other efficacy secondary endpoints showed trends in favour of THC:CBD spray but
without reaching statistical significance. This might relate to the study power (the sample size was
calculated for the primary endpoint) and also to the parameters per se and the nature of MS
spasticity as being one of many symptoms of MS [3].
Mean daily use of THC:CBD spray was 7.7 sprays/day at the end of Phase A and 7.3 sprays/day
(versus 8.5 sprays/day for placebo) at the end of 12 weeks’ treatment in Phase B. This level of usage
is lower than that reported in the largest pivotal phase 3 clinical trial of THC:CBD spray (8.3
sprays/day) [16], and consistent with that reported in observational studies in daily practice (6.7-6.8
sprays/day) [17,18]. Evidence is accumulating to suggest that patients receiving THC:CBD spray in
daily practice gain sufficient benefit at a mean dosage of about 7 sprays/day.
THC:CBD spray was well tolerated during both the trial and treatment phases of the study. In line
with observational studies of THC:CBD spray [17,18], incidences of treatment-related AEs were low
overall and any AEs reported were mainly mild or moderate in intensity and not different from those
reported in the previous studies (e.g. dizziness, somnolence). The incidence of AEs related to
THC:CBD spray decreased from 19.4% of patients in Phase A to 9.4% of patients in Phase B, likely
reflecting accustomisation by patients during continued use, which was further supported by a
decrease in the incidence of treatment interruptions related to AEs in THC:CBD spray-treated
patients, from 15.2% of patients in Phase A to 3.8% of patients in Phase B.
The design of this study, especially the inclusion of a washout period to avoid carryover and other
effects of THC:CBD spray during the randomised treatment phase, allowed us to better evaluate its
true therapeutic effect. Under these conditions, THC:CBD spray at an average dose of about 7
sprays/day produced clinically meaningful improvement of resistant MS spasticity in more than
three-quarters of initial responders, thus proving to be a more efficacious alternative to readjusting
underlying antispasticity drugs.
Acknowledgements
The authors wish to express their sincere thanks for the collaboration of participating patients and
investigators *CZ: Y. Benešová (Brno), M. Dufek (Brno), P. Hradílek (Havířov), L. Janů (Plzeň), H.
Lachmann (Prague), B. Luběnová (Olomouc), E. Meluzínová (Prague), I. Nováková (Prague), O. Škoda
(Pelhřimov), I. Štětkářová (Prague), R. Taláb (Hradec Králové), M. Vachová (Teplice), M. Vališ
(Choceň); AT: M. Guger (Linz)+, all team members at study sites, Easthorn CRO, and Almirall S.A.
Writing assistance was provided by Content Ed Net (Madrid, Spain), with funding from Almirall S.A.
(Barcelona, Spain).
Disclosure statement
J Markovà has received fees as consultant or investigator in projects sponsored by Almirall, Amgen,
Biogen, Merck, Novartis, Pfizer, Teva and other pharmaceutical companies.
U Essner and A Lenschat are consultants for Almirall Hermal GmbH.
B Akmaz and C Trompke are fulltime employees of Almirall Hermal GmbH.
M Marinelli and C Vila are fulltime employees of Almirall S.A.
Funding details
This study was sponsored by Almirall Hermal GmbH and Almirall S.A.
References
1. Rizzo MA, Hadjimichael OC, Preiningerova J, et al. Prevalence and treatment of spasticity
reported by multiple sclerosis patients. Mult Scler. 2004;10:589595.
2. de Sa JC, Airas L, Bartholome E, et al. Symptomatic therapy in multiple sclerosis: a review for a
multimodal approach in clinical practice. Ther Adv Neurol Disord. 2011;4:139-168.
3. Kister I, Bacon TE, Chamot E, et al. Natural history of multiple sclerosis symptoms. Int J MS Care.
2013;15:146158.
4. Gold R, Oreja-Guevara C. Advances in the management of multiple sclerosis spasticity: multiple
sclerosis spasticity guidelines. Expert Rev Neurother. 2013;13(12 Suppl):5559.
5. Milinis K, Tennant A, Young CA, et al. Spasticity in multiple sclerosis: Associations with
impairments and overall quality of life. Mult Scler Relat Disord. 2016;5:3439.
6. Svensson J, Borg S, Nilsson P. Costs and quality of life in multiple sclerosis patients with
spasticity. Acta Neurol Scand. 2014; 129: 1320.
7. Flachenecker P, Henze T, Zettl UK. Spasticity in patients with multiple sclerosis-clinical
characteristics, treatment and quality of life. Acta Neurol Scand. 2014; 129: 154162.
8. Arroyo R, Massana M, Vila C. Correlation between spasticity and quality of life in patients with
multiple sclerosis: the CANDLE study. Int J Neurosci 2013; 123: 850858.
9. Oreja-Guevara C, Montalban X, de Andrés C, et al.; Grupo de Enfermedades Desmielinizantes de
la Sociedad Española de Neurología. [Consensus document on spasticity in patients with multiple
sclerosis. Grupo de Enfermedades Desmielinizantes de la Sociedad Española de Neurología]. Rev
Neurol 2013; 57(8):359-73.
10. Beard S, Hunn A, Wight J. Treatments for spasticity and pain in multiple sclerosis: a systematic
review. Health Technol Assess. 2003;7(40):iii, ix-x, 1-111.
11. Chang E, Ghosh N, Yanni D, et al. A Review of Spasticity Treatments: Pharmacological and
Interventional Approaches. Crit Rev Phys Rehabil Med. 2013;25:1122.
12. Pozzilli C. Advances in the management of multiple sclerosis spasticity: experiences from recent
studies and everyday clinical practice. Expert Rev Neurother. 2013;13(12 Suppl):4954.
13. Otero-Romero S, Sastre-Garriga J, Comi G, et al. Pharmacological management of spasticity in
multiple sclerosis: Systematic review and consensus paper. Mult Scler. 2016;22:1386-1396.
14. Wade DT, Makela P, Robson P, et al. Do cannabis-based medicinal extracts have general or
specific effects on symptoms in multiple sclerosis? A double-blind, randomized, placebo-
controlled study on 160 patients. Mult Scler. 2004;10:434441.
15. Collin C, Davies P, Mutiboko IK, et al. Randomized controlled trial of cannabis-based medicine in
spasticity caused by multiple sclerosis. Eur J Neurol. 2007;14:290296.
16. Novotna A, Mares J, Ratcliffe S, et al. A randomized, double-blind, placebo-controlled, parallel-
group, enriched-design study of nabiximols* (Sativex(®) ), as add-on therapy, in subjects with
refractory spasticity caused by multiple sclerosis. Eur J Neurol. 2011;18:11221131.
17. Flachenecker P, Henze T, Zettl UK. Nabiximols (THC/CBD oromucosal spray, Sativex ®) in clinical
practice results of a multicenter, non-interventional study (MOVE 2) in patients with multiple
sclerosis spasticity. Eur Neurol. 2014;71:271279.
18. Patti F, Messina S, Solaro C, et al. Efficacy and safety of cannabinoid oromucosal spray for
multiple sclerosis spasticity. J Neurol Neurosurg Psychiatry. 2016;87:944951.
19. eMC Sativex Oromucosal Spray SPC. 2015; [cited 2017 Sep 22] Available from:
https://www.medicines.org.uk/emc/history/23262
20. Farrar JT, Troxel AB, Stott C, et al. Validity, reliability, and clinical importance of change in a 0-10
numeric rating scale measure of spasticity: a post hoc analysis of a randomized, double-blind,
placebo-controlled trial. Clin Ther. 2008;30:974985.
21. Collin C, Ehler E, Waberzinek G, et al. A double-blind, randomized, placebo-controlled, parallel-
group study of Sativex, in subjects with symptoms of spasticity due to multiple sclerosis. Neurol
Res. 2010;32(5):451459.
22. Wininger M, Craelius W, Settle J, et al. Biomechanical analysis of spasticity treatment in patients
with multiple sclerosis. Ther Adv Neurol Disord. 2015;8(5):203211.
Figures
Figure 1. Overview of study design. Non-returners were initial responders who failed to show a ≥
80% reduction of their Phase A NRS improvement during washout.
Figure 2. Patient disposition. Abbreviations: IMP: Investigational Medicinal Product; NRS, Numerical
Rating Scale.
Figure 3. Primary endpoint: proportion of MS spasticity 0-10 NRS CID responders (≥ 30%
improvement from baseline) after 12 weeks’ randomised double-blind treatment with THC:CBD
oromucosal spray or placebo. CI, Confidence interval; CID, Clinically important difference; NRS,
Numerical rating scale. OR, Odds ratio.
Figure 4. Change in MS spasticity 0-10 NRS score (measured in Least Squares [LS] means) from Phase
B baseline (week 1) through to week 12 of randomised double-blind treatment with THC:CBD
oromucosal spray (n = 53) or placebo (n = 53) in the intent-to-treat population.
Tables
Table 1. Demographic and clinical characteristics of patients entering the Phase A trial
period (n = 191).
Characteristic
Value
Gender: female/male: n (%)
134 (70.2%) / 57 (29.8%)
Age: mean (SD)
51.3 (10.2)
MS classification: n (%)
Primary progressive MS
Secondary progressive MS
Relapsing remitting MS
21 (11.0%)
92 (48.2%)
78 (40.8%)
MS disease history, years: mean (SD)
14.2 (8.4)
EDSS score: mean (SD)
5.9 (1.1)
Disease modifiers use n (%):
Past
Present
191 (100%)
190 (99.5%)
MS spasticity history, years: mean (SD)
7.8 (5.3)
NRS spasticity score: mean (SD)
6.4 (1.2)
Pain NRS: mean (SD)
5.5 (1.9)
Table 2. Secondary efficacy endpoints: least squares (LS) means for change in measures of spasticity
and associated symptoms from Phase B baseline to week 12.
Endpoint
THC:CBD
oromucosal spray
(mean) (n = 53)
Placebo
(mean)
(n = 53)
P-value
Spasticity NRS score, 0-10
scale
-3.5
-1.6
< 0.0001
Pain NRS score, 0-10 scale
-3.2
-1.8
< 0.0013
MAS score, 6-point ordinal
scale
-0.30
-0.06
< 0.0007
MS EDSS score, 0-10 scale
-0.02
-0.01
< 0.6529
Spasms frequency, n
-20.58
-17.75
ns
Spasms severity, 0-3 scale
-0.72
-0.38
0.0001
Sleep disruption NRS score, 0-
10 scale
-3.21
-1.78
0.0006
MAS score per muscular
group, 0-4 scale
Elbow flexor
Elbow extensor
Elbow pronator
Elbow supinator
Wrist flexor
Finger flexor
Hip adductor
Knee flexor
Knee extensor
-0.28
-0.31
-0.23
-0.16
-0.16
-0.16
-0.43
-0.47
-0.51
-0.33
-0.03
-0.01
-0.02
-0.02
-0.04
-0.02
-0.09
-0.11
-0.13
-0.10
0.0044
0.0004
0.0327
ns
ns
ns
0.0007
0.0016
0.0004
0.0119
Foot plantar
Barthel ADL index
0.04
0.11
ns
SF-36 (General Health)
0.31
1.90
ns
Timed 10-meter walk,
seconds
-2.79
-1.08
0.11
SGIC, adjusted odds ratio and
P-value THC:CBD vs. placebo
Week 4 2.852, P=0.0035 Week 8 1.823, P=0.1331
Week 12 1.384, P=0.3515
PGIC, adjusted odds ratio and
P-value THC:CBD vs. placebo
Week 4 3.972, P=0.00005 Week 8 2.418, P=0.0260
Week 12 1.623, P=0.1615
ADL, Activities of daily living; EDSS: Expanded Disability Status Scale; MAS: modified Ashworth scale;
MS, Multiple sclerosis; NRS: Numerical rating scale; ns, Not significant; PGIC, Physician’s global
impression of change; SF-36: 36-Item Short Form health survey; SGIC: Subject’s/patient’s global
impression of change.
Table 3: Treatment-emergent adverse events (TEAEs) in the Phase A and washout safety population.
Total (n = 191)
n (%)
Events
Phase A
Serious adverse events (SAEs)
2 (1.0%)
3
Adverse events (AEs)
46 (24.1%)
75
Relationship to IMP
Not related to IMP
9 (4.7%)
11
Related to IMP
37 (19.4%)
64
Intensity
Mild
34 (17.8%)
47
Moderate
13 (6.8%)
28
Number of treatment interruptions*
related to AEs
29 (15.2%)
54
Adverse events leading to withdrawal
4 (2.1%)
5
Adverse events leading to death
0
0
Washout
SAEs
0
0
AEs
12 (6.3%)
20
Relationship to IMP
Not related
6 (3.1%)
8
Related
6 (3.1%)
12
Intensity
Mild
6 (3.1%)
10
Moderate
6 (3.1%)
10
Number of treatment interruptions*
related to AEs
6 (3.1%)
12
Adverse events leading to withdrawal
1 (0.5%)
1
Adverse events leading to death
0
0
IMP: Investigational medicinal product.
*Treatment interruptions includes the following action taken with study treatment: drug withdrawn,
dose reduced, or dose increased.
An adverse event that has a start and stop date in different study phases has been presented in all
phases that the event was ongoing.
Table 4. Treatment-emergent adverse events (TEAEs) during the 12-week double-blind treatment
period (Phase B), in the Phase B safety population.
THC:CBD oromucosal
spray
(n = 53)
Placebo
(n = 53)
n (%)
Events
n (%)
Events
Serious adverse events
(SAEs)
1 (1.9%)
1
1 (1.9%)
1
Adverse events (AEs)
including SAEs
12 (22.6%)
19
7 (13.2%)
8
Relationship to IMP
Not related to IMP
8 (15.1%)
12
6 (11.3%)
7
Related to IMP
5 (9.4%)
7
1 (1.9%)
1
Intensity
Moderate
6 (11.3%)
6
0
0
Mild
7 (13.2%)
13
6 (11.3%)
7
Severe
0
0
1 (1.9%)
1
Number of treatment
interruptions related to
AEs
2 (3.8%)
3
0
0
IMP: Investigational medicinal product.
*Treatment interruptions includes the following action taken with study treatment: drug withdrawn,
dose reduced, or dose increased.
An adverse event that has a start and stop date in different study phases has been presented in all
phases that the event was ongoing.
... Optimization of the underlying antispasticity medications was permitted in both groups across all study periods(Markovà et al., 2019). The study criteria included adult (18y+) multiple sclerosis patients that had encountered spasticity for at least 12 months and had co-existing symptoms of chronic pain, spasm, low mobility, sleep disturbance, depression, anxiety, and fatigue(Markovà et al., 2019). ...
... Optimization of the underlying antispasticity medications was permitted in both groups across all study periods(Markovà et al., 2019). The study criteria included adult (18y+) multiple sclerosis patients that had encountered spasticity for at least 12 months and had co-existing symptoms of chronic pain, spasm, low mobility, sleep disturbance, depression, anxiety, and fatigue(Markovà et al., 2019). The criteria excluded who had taken THC, CBD spray in the past, or currently using cannabis or cannabinoid-based drugs of any form within 30 days prior to the study period and any family history of psychiatric disorders(Markovà et al., 2019).MM treatments for QOL and SEWB An 8-week prospective, single-arm cohort study of 97 chronic pain patients for the potential role of cannabinoids as an alternative for opioids concluded a significant effect on reducing opioid intake, reducing pain, and improving QOL(Capano et al., 2020). ...
... The study criteria included adult (18y+) multiple sclerosis patients that had encountered spasticity for at least 12 months and had co-existing symptoms of chronic pain, spasm, low mobility, sleep disturbance, depression, anxiety, and fatigue(Markovà et al., 2019). The criteria excluded who had taken THC, CBD spray in the past, or currently using cannabis or cannabinoid-based drugs of any form within 30 days prior to the study period and any family history of psychiatric disorders(Markovà et al., 2019).MM treatments for QOL and SEWB An 8-week prospective, single-arm cohort study of 97 chronic pain patients for the potential role of cannabinoids as an alternative for opioids concluded a significant effect on reducing opioid intake, reducing pain, and improving QOL(Capano et al., 2020). The study enrolled 30-65 year old patients suffering from moderate to severe chronic pain for at least 3 years and had been stable on opioids for 1 year. ...
Article
Background Recent studies have established that Medicinal Marijuana (MM) is beneficial in the treatment of spasms, sleep, and pain in adult patients with varying medical diagnoses and symptoms. However, MM has rarely been used for the treatment of Cerebral Palsy (CP) complications in adults. The aim of this systematic literature review was to explore MM interventions globally, with a focus on identifying the best practice with MM for the treatment of complications of CP. Methods A systematic literature search was performed using keywords and synonyms related to MM treatment and CP complications. Inclusions and exclusions were scoped to scholarly peer reviewed academic literature published 2019 to 2021 located in the Deakin Library collection. A screening process confirmed criteria adherence and identified additional papers in referencing. The papers were appraised and evaluated to ensure selections do not have perceived or actual bias. Results From 409 publications, 27 papers were selected for review because they investigated the benefits of MM treatment for patients with sleep, pain, and spasm complications. There was no literature found on the use of MM for adults with CP. Discussion Recent research has demonstrated that with an informed understanding of MM treatment adult patients with varying medical diagnoses and symptoms can use MM to manage sleep disruption and improve relaxation. Therefore, there are potential benefits for the use of MM in treating spasticity, pain, sleep, quality of life, and social and emotional wellbeing in adult patients with CP. No funding was sort or provided for this review and the results are specific to adults with CP, so they are not to be generalized to other populations.
... Cannabinoids can also be used to alleviate rheumatoid arthritis by regulating the immune response [34]. CBD and THC ameliorate autoimmune effect on the nervous system, and are, hence, used to treat severe spasticity and pain in MS [57]. They have also demonstrated potential in the treatment of systemic lupus erythematosus [58], type 1 diabetes [59], and various types of neurological disorders, e.g., Alzheimer's disease [60]. ...
... Sativex is the trademark of a cannabis drug called Nabiximols consisting of THC (2.7 mg per dose) and CBD (2.5 mg per dose). It has already been approved in Canada and most European countries for the treatment of moderate to severe spasticity in MS [57]. The license authorizes its use as a complement for treating MS. ...
Article
Full-text available
Cannabis sativa L. contains numerous compounds with antioxidant and anti-inflammatory properties, including the flavonoids and the cannabinoids, particularly Δ-9-tetrahydrocannabinol (THC) and cannabidiol (CBD). Cannabinoids have an effect on the endocannabinoid system (ECS), a cellular communication network, and are, hence, widely studied for medical applications. Epidiolex®, a 99% pure oral CBD extract, has been approved by the FDA for the treatment of epilepsy. Nabiximols (Sativex) is an oromucosal spray containing equal volume of THC and CBD, and it is commonly used as an add-on treatment for unresponsive spasticity in multiple sclerosis (MS) patients. Several in vitro and in vivo studies have also shown that cannabinoids can be used to treat various types of cancer, such as melanoma and brain glioblastoma; the first positive clinical trials on the anticancer effect of a THC:CBD blend with temozolomide (TMZ) in the treatment of highly invasive brain cancer are very promising. The cannabinoids exert their anticancer properties in in vitro investigations by the induction of cell death, mainly by apoptosis and cytotoxic autophagy, and the inhibition of cell proliferation. In several studies, cannabinoids have been found to induce tumor regression and inhibit angiogenic mechanisms in vitro and in vivo, as well as in two low-numbered epidemiological studies. They also exhibit antiviral effects by inhibiting ACE2 transcription, blocking viral replication and fusion, and acting as anti-inflammatory agents; indeed, prior CBD consumption (a study of 93,565 persons in Chicago) has also been associated with a much lower incidence of SARS-CoV-2 infections. It is postulated that cannabis extracts can be used in the treatment of many other diseases such as systemic lupus erythematosus, type 1 diabetes, or various types of neurological disorders, e.g., Alzheimer’s disease. The aim of this review is to outline the current state of knowledge regarding currently used medicinal preparations derived from C. sativa L. in the treatment of selected cancer and viral diseases, and to present the latest research on the potential applications of its secondary metabolites.
... The successful use of cannabinoids to alleviate spasticity in adult patients with multiple sclerosis (MS) patients [10,21], as evidenced by the approval of Nabiximols (a Cannabis sativa extract; CBD:THC ratio of 1:1), has led to multiple efforts to develop cannabinoid formulations that would similarly benefit paediatric patients [5][6][7][8]11,13,22]. Given the role of the endocannabinoid system [23], a certain amount of THC is believed to be necessary to relieve spasticity. ...
... Drowsiness, which is a frequently described AE of THC-containing cannabis preparations [15,22], was also the most Table 3 Summary of adverse events affecting more than five out of 60 participants, expressed as the number of patients affected by each AE. [18][19][20][21][22][23][24] common AE in our trial, whereas Fairhurst did not describe this AE at all [13]. Unlike Fairhurst [13], we did not observe hallucinogenic properties, while some parents in our trial described episodes of laughter and inappropriate behaviour in their children. ...
... In rodent studies in a multiple sclerosis model, desirable effects were obtained after the simultaneous administration of CBD and THC compared to these substances administered separately [75]. Some human studies confirm the positive effect of combining CBD and THC in reducing spasticity in people with multiple sclerosis [3,63,76]. Their potential may also be related to reducing pain associated with multiple sclerosis, but further clinical trials on large populations are also required in this regard [63,75]. ...
Article
Full-text available
This review provides an overview of cannabis-based phytocannabinoids, focusing on their mechanisms of action, therapeutic applications, and production processes, along with the environmental factors that affect their quality and efficacy. Phytocannabinoids such as THC (∆9-tetrahydrocannabinol), CBD (cannabidiol), CBG (cannabigerol), CBN (cannabinol), and CBC (cannabichromene) exhibit significant therapeutic potential in treating various physical and mental health conditions, including chronic pain, epilepsy, neurodegenerative diseases, skin disorders, and anxiety. The cultivation of cannabis plays a crucial role in determining cannabinoid profiles, with indoor cultivation offering more control and consistency than outdoor methods. Environmental factors such as light, water, temperature, humidity, nutrient management, CO2, and the drying method used are key to optimizing cannabinoid content in inflorescences. This review outlines the need for broader data transfer between the health industry and technological production, especially in terms of what concentration and cannabinoid ratios are effective in treatment. Such data transfer would provide cultivators with information on what environmental parameters should be manipulated to obtain the required final product.
... Patients were defined as responders to nabiximols in case of an improvement of at least 30% in the sNRS score after 12 weeks of treatment [19,21]. In a further analysis, we defined nabiximols responders as those patients improving by at least 30% in the pNRS score after 12 weeks of treatment, pain being the most frequent and invalidating spasticity-related symptom [8]. ...
Article
Full-text available
Background: Cannabinoid oro-mucosal spray nabiximols is approved for patients with moderate to severe multiple sclerosis spasticity (MSS) resistant to other antispastic medications. Few real-world data are available on the effectiveness, safety and patients’ satisfaction in MS patients treated with nabiximols as monotherapy. Methods: To investigate the effectiveness, tolerability and satisfaction of nabiximols in a real-life multicentric Swiss cohort as monotherapy or with stable doses of other antispastic medications, and explore clinical features which may predict treatment response. The following data were collected at treatment start (baseline) and 12 weeks thereafter: Modified Ashworth scale (MAS), scores at numerical rating scales ranging from 0 (absent) to 10 (considerable) for effect on spasticity (sNRS), pain (pNRS), gait (gNRS), urinary symptoms (uNRS), tolerability (tNRS) as assessed by the treating neurologist, and overall treatment satisfaction (TsNRS) and tolerability (tNRS) as assessed by the patient. Results: Ninety-five patients (44 relapsing remitting, 37 secondary progressive and 14 primary progressive MS; median age = 53 (IQR 45–62); female 70%; median EDSS 6 (IQR 4–6), concomitant antispastic treatments in 54% of patients) were included. From baseline to week 12, median MAS score decreased from 3.0 to 2.0 (p < 0.001). Median scores of the each NRS also significantly decreased (p < 0.001 for all comparisons). At week 12, the median TsNRS and tTS scores were 8/10 (IQR: 6–9) and 9/10 (IQR: 7–10), respectively, and 93.7% of patients continued to use nabiximols at the average dose of six sprays/day. No clinical factors, including use of nabiximols as add on vs. monotherapy, were associated with responder status. Conclusions: Our first Swiss, multicentric, observational, real-life study supports and enhances previous finding of nabiximols as monotherapy and as add-on therapy, being an effective, safe and well-tolerated treatment option for resistant MS spasticity and spasticity-related symptoms (pain, bladder dysfunction and gait).
Chapter
Multiple sclerosis (MS) is a complex autoimmune disease characterized by inflammation, demyelination, and neurodegeneration, with oxidative stress playing a pivotal role in its pathogenesis. Recognizing the importance of mitigating oxidative damage, this abstract provides a succinct overview of the therapeutic potential of specific antioxidants—Ginkgo biloba, melatonin, curcumin, Edaravone, and N-acetyl cysteine—in the context of MS. Ginkgo biloba, a renowned herbal supplement, possesses robust antioxidant and anti-inflammatory properties. Preclinical phase studies suggest it can modulate immune responses, reduce neuroinflammation, and potentially impede MS progression. Melatonin, a potent antioxidant hormone, displays neuroprotective and immunomodulatory effects, positioning it as a promising adjunct therapy for MS. Curcumin, the active component of turmeric, demonstrates anti-inflammatory and antioxidant actions, offering potential benefits in reducing neuroinflammation and promoting remyelination. Edaravone, a free radical scavenger approved for neurodegenerative disorders, holds promise in alleviating MS-related oxidative stress. N-acetyl cysteine, a glutathione precursor, exhibits neuroprotective and anti-inflammatory properties, making it a potential therapeutic avenue for MS. This chapter will delve into the therapeutic potential of these antioxidants for MS management through a discussion of preclinical and clinical trials.
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
Full-text available
Interest in medical cannabis and cannabis-based medicinal products (CBMPs) has increased greatly in recent years. Two cannabinoids are of principal importance; delta-9-tetrahydrocannabinol (∆9-THC), the primary psychoactive component, and also cannabidiol (CBD), considered non-intoxicating. Each has distinct mechanisms of action and different therapeutic potentials. CBMPs differ in their ∆9-THC and CBD components; predominantly ∆9-THC, balanced formulations with equivalent ∆9-THC and CBD elements, and CBD-predominant products. In this narrative review, we evaluate the published evidence for the clinical benefits of CBMPs and overall benefits in well-being. We also review the overall safety profile and discuss the potential for dependence with CBMPs. Evidence can be drawn from a wide range of randomized and other controlled studies and from observational real-world studies. Most data from observational registry studies are supportive of ∆9-THC-based products (∆9-THC-predominant or balanced CBMPs) in the management of chronic neuropathic pain. Balanced products are also effective in reducing spasticity in multiple sclerosis. Most CBMPs show benefit in providing symptomatic benefits in reducing anxiety, nausea, and in improving sleep, but the place of specific products is more subtle, and choice guided by specific circumstances. Symptomatic improvements are accompanied by improved quality of life and well-being. Safety data indicate that CBMPs are generally well tolerated in most patients without specific contraindications. The majority of adverse effects are non-serious, and transient; most are principally associated with ∆9-THC and are dose-dependent. In contrast to recreational cannabis use, there is little evidence from clinical studies that CBMPs have any potential for dependence.
Article
Full-text available
Cannabinoids are compounds found in and derived from the Cannabis plants that have become increasingly recognised as significant modulating factors of physiological mechanisms and inflammatory reactions of the organism, thus inevitably affecting maintenance of homeostasis. Medical Cannabis popularity has surged since its legal regulation growing around the world. Numerous promising discoveries bring more data on cannabinoids’ pharmacological characteristics and therapeutic applications. Given the current surge in interest in the medical use of cannabinoids, there is an urgent need for an effective method of their administration. Surpassing low bioavailability, low water solubility, and instability became an important milestone in the advancement of cannabinoids in pharmaceutical applications. The numerous uses of cannabinoids in clinical practice remain restricted by limited administration alternatives, but there is hope when biodegradable polymers are taken into account. The primary objective of this review is to highlight the wide range of indications for which cannabinoids may be used, as well as the polymeric carriers that enhance their effectiveness. The current review described a wide range of therapeutic applications of cannabinoids, including pain management, neurological and sleep disorders, anxiety, and cancer treatment. The use of these compounds was further examined in the area of dermatology and cosmetology. Finally, with the use of biodegradable polymer-based drug delivery systems (DDSs), it was demonstrated that cannabinoids can be delivered specifically to the intended site while also improving the drug’s physicochemical properties, emphasizing their utility. Nevertheless, additional clinical trials on novel cannabinoids’ formulations are required, as their full spectrum therapeutical potential is yet to be unravelled.
Article
Full-text available
Background The approval of 9-δ-tetrahydocannabinol and cannabidiol (THC:CBD) oromucosal spray (Sativex) for the management of treatment-resistant multiple sclerosis (MS) spasticity opened a new opportunity for many patients. The aim of our study was to describe Sativex effectiveness and adverse events profile in a large population of Italian patients with MS in the daily practice setting. Methods We collected data of all patients starting Sativex between January 2014 and February 2015 from the mandatory Italian medicines agency (AIFA) e-registry. Spasticity assessment by the 0–10 numerical rating scale (NRS) scale is available at baseline, after 1 month of treatment (trial period), and at 3 and 6 months. Results A total of 1615 patients were recruited from 30 MS centres across Italy. After one treatment month (trial period), we found 70.5% of patients reaching a ≥20% improvement (initial response, IR) and 28.2% who had already reached a ≥30% improvement (clinically relevant response, CRR), with a mean NRS score reduction of 22.6% (from 7.5 to 5.8). After a multivariate analysis, we found an increased probability to reach IR at the first month among patients with primary and secondary progressive MS, (n=1169, OR 1.4 95% CI 1.04 to 1.9, p=0.025) and among patients with >8 NRS score at baseline (OR 1.8 95% CI 1.3–2.4 p<0.001). During the 6 months observation period, 631(39.5%) patients discontinued treatment. The main reasons for discontinuation were lack of effectiveness (n=375, 26.2%) and/or adverse events (n=268, 18.7%). Conclusions Sativex can be a useful and safe option for patients with MS with moderate to severe spasticity resistant to common antispastic drugs.
Article
Full-text available
Objectives: New metrics for clinical spasticity are needed to assess motor performance, since scales such as the Ashworth and Tardieu are unreliable. Here, we assessed outcomes of baclofen treatment in patients with multiple sclerosis (MS) using biomechanical analysis of voluntary movements. Methods: Patients with MS and symptomatic limb spasticity were recruited for a pre-post baclofen titration study, along with age-matched healthy controls. Oral baclofen was titrated to optimize spasticity symptoms in all MS cases over 4 weeks. Clinical assessments included the Modified Ashworth Scale (MAS), Tardieu Scale (TS); elbow kinematics were measured via the Transient Acceleration Measurement Interface (TAMI); performance was measured as the score at 4 weeks minus the baseline score in all measures. Movement proficiency within TAMI was quantified through a scale-free smoothness measure, according to the regional excursion deviation (RED) from a constant-velocity approximant. Results: Twelve patients with MS [age: 47.8 ± 9.8 years; women: 4; disease duration: 20 ± 10 years; disease-modifying therapy use: 7; Expanded Disability Status Scale (EDSS): 6.8 ± 1.4] and eight age-matched healthy controls were evaluated concurrently (mean age: 49.5 ± 13.1 years; women = 3). In MS cases, no significant improvement in arm spasticity was observed with main effects: MAS: -41.6 ± 72.6 (p = 0.09); EDSS: -1.6 ± 10.4% (p = 0.49); and TS: -8.3 ± 2.1% (p = 0.32), -24.9 ± 63.6% (p = 0.42), and -30.7 ± 79.9% (p = 0.06), at slow, moderate, and fast speeds, respectively. However, voluntary motion smoothness, as measured by TAMI: RED, decreased significantly: 0.62 ± 0.08 versus 0.54 ± 0.09, p < 0.001, indicating significant increase in movement smoothness post treatment. Conclusion: A simple biomechanical analysis of voluntary movements revealed a significant reduction of spasticity after 30 days of baclofen therapy in patients with MS that was not detected by clinical assessments.
Article
Full-text available
The North American Research Committee on Multiple Sclerosis (NARCOMS) Registry is a database that contains information from over 35,000 patient volunteers on symptom severity in 11 domains commonly affected in multiple sclerosis (MS): mobility, hand function, vision, fatigue, cognition, bowel/bladder function, sensory, spasticity, pain, depression, and tremor/coordination. The Registry affords a unique opportunity to study the frequency and severity of domain-specific impairment in a contemporary, mostly treated MS cohort over the course of the disease. The objective of this work was to calculate symptom prevalence in each of the 11 domains for years 0 to 30 from symptom onset. The resulting "symptom prevalence tables" demonstrate that a majority of participants perceive at least some degree of impairment in most domains as early as the first year of disease. The severity of impairment increases with disease duration across all domains, but the patterns of disability accumulation differ. The symptom prevalence tables illustrate the magnitude of perceived impact of the disease and highlight the extent of unmet need in symptomatic management. The tables are easy to use and allow MS patients and their clinicians to compare an individual's own impairment in any of the 11 domains to that of NARCOMS participants with the same disease duration.
Article
Full-text available
Symptomatic therapy of multiple sclerosis (MS) is an important part of a comprehensive treatment plan that aims to improve patients' quality of life. In the current era of medical progress, several factors have led to the development of guidelines for MS management. There is continued need for an evidence-based approach supported by high-quality data from controlled clinical trials. Most healthcare systems require this approach and include it in the reimbursement process. Guidelines are usually committed by national or continental neurological societies. The Spanish Society of Neurology demyelinating diseases working group has developed a consensus document on spasticity in patients with MS. MS experts from the group used the metaplan method to sum up the most important recommendations about spasticity for inclusion in the guidance. Recommendations were classified according to the Scottish Intercollegiate Guidelines Network system and approved by all members of the group. In Germany, the guideline panel of the German Neurological Society endorsed the national competence network for multiple sclerosis (Krankheitsbezogenes Kompetenznetz Multiple Sklerose) to update the existing recommendations. The most recent fifth edition of the guidelines (dated April 2012) now also includes recommendations for treatment of key symptoms such as spasticity. More than 30 MS neurologists contributed to the new edition reflecting the need for broad expertise. After a first round in which key topics were defined, a web-based decision process was undertaken to further develop individual topics such as symptomatic therapy. The draft manuscript was reviewed once again by the group prior to submission to the official review process. The aims of spasticity treatment are to improve mobility and dexterity, achieve physiological movement patterns, reduce pain, facilitate nursing measures and avoid complications such as contractures. Representative antispasticity medications include baclofen, tizanidine, gabapentin, dantrolene, tolperisone, benzodiazepines and Sativex® oromucosal spray. Botulinum toxin and intrathecal baclofen may also be required in selected cases. Plans are currently in motion to develop next-level European guidelines through a concerted approach coordinated by the European Federation of Neurological Societies.
Article
Background and objectives: Treatment of spasticity poses a major challenge given the complex clinical presentation and variable efficacy and safety profiles of available drugs. We present a systematic review of the pharmacological treatment of spasticity in multiple sclerosis (MS) patients. Methods: Controlled trials and observational studies were identified. Scientific evidence was evaluated according to pre-specified levels of certainty. Results: The evidence supports the use of baclofen, tizanidine and gabapentin as first-line options. Diazepam or dantrolene could be considered if no clinical improvement is seen with the previous drugs. Nabiximols has a positive effect when used as add-on therapy in patients with poor response and/or tolerance to first-line oral treatments. Despite limited evidence, intrathecal baclofen and intrathecal phenol show a positive effect in severe spasticity and suboptimal response to oral drugs. Conclusion: The available studies on spasticity treatment offer some insight to guide clinical practice but are of variable methodological quality. Large, well-designed trials are needed to confirm the effectiveness of antispasticity agents and to produce evidence-based treatment algorithms.
Article
Objectives: Design: Cross-sectional survey. Setting: A convenience sample of people with MS routinely attending an appointment with their local MS service. Participants: 701 patients with clinically definite MS. Main outcome measures: Demographic details were obtained and patients completed a battery of measures including spasticity (Multiple Sclerosis Spasticity Scale - 88), fatigue (Neurological Fatigue Index - MS), urinary dysfunction (Qualiveen-SF), pain (Neuropathic Pain Scale), mood disorder (Hospital Anxiety and Depression Scale), disability (World Health Organisation Disability Assessment Schedule) and QOL (Leeds Multiple Sclerosis QOL Scale). Results: 85.7% of patients reported spasticity. Patients with higher levels of spasticity were more likely to be disabled, suffer from depression and anxiety, have higher levels of fatigue and report more pain and bladder problems (p<0.01). Spasticity remained as a significant direct effect upon QOL in a multivariate model adjusted for other impairments, activity limitation and depression. Conclusions: There is a strong association between spasticity and fatigue, depression, anxiety, pain and bladder problems. The retention of a significant direct relationship with QOL in a multivariate model emphasises its influence upon the everyday lives of people with MS.
Article
Spasticity is a velocity-dependent increase in muscle tone and uncontrolled, repetitive, involuntary contractions of skeletal muscles. Spasticity presents as upper motor neuron symptoms in patients with central nervous system pathology such as stroke, spinal cord injury, brain injury, or multiple sclerosis. As a result, a patient can have significant pain and limited mobility, which can lead to decreased quality of life and difficulty maintaining personal care. In this article we discuss mechanisms, indications, efficacy, and side effects of the most accepted current treatments. Currently available treatment options include oral medications and interventional procedures. Oral medications comprise centrally acting agents, such as baclofen, clonidine, and tizanidine, as well as anticonvulsants such as benzodiazepines and gabapentin and peripherally acting dantrolene. Interventional procedures include focal injections of botulinum toxin, phenol or alcohol, and an intrathecal baclofen pump. Surgical treatments include selective dorsal rhizotomy and neurectomy. We found that there are several treatments available with data to support their use, but many still need further research to prove their efficacy and develop optimal utilization.
Article
Background: Nabiximols (Sativex®), a cannabinoid-based oromucosal spray, is an add-on therapy for patients with moderate to severe multiple sclerosis spasticity (MSS) resistant to other medications. The primary objective was to provide real-life observational data of clinical experience of nabiximols in contrast to formal clinical trials of effectiveness. Methods: This was an observational, prospective, multicenter, non-interventional study with a follow-up period of 3-4 months, conducted in routine care setting in Germany. Patients with moderate to severe MSS were included at nabiximols' initiation. Structured documentation forms, questionnaires and validated instruments were used for data collection at inclusion, 1 and 3 months after inclusion. Results: Overall, 335 patients were assessed of whom 276 fitted the criteria and were included in the effectiveness analysis. After 1 month, nabiximols provided relief of resistant MSS in 74.6% of patients according to specialist assessment; mean spasticity 0-10 numerical rating scale (NRS) score decreased from 6.1 ± 1.8 to 5.2 ± 2.0 points; in patients with NRS improvement ≥20% mean NRS score decreased by 40%. After 3 months, 55.3% of patients had continued to use nabiximols and the mean NRS score had decreased by 25% from baseline. 17% of patients reported adverse events. Conclusion: Real-life data confirm nabiximols as an effective and well-tolerated treatment option for resistant MSS in clinical practice.
Article
Although spasticity of varying severity affects up to 80% of patients with multiple sclerosis (MS) during the course of their disease, the symptom is often overlooked and undertreated. Despite the availability of oral antispasticity treatments (baclofen, tizanidine and others), approximately one-third of MS patients in Europe and the USA experience moderate or severe nonfocalized spasticity. At present, a thorough clinical evaluation of MS-related spasticity that takes into account the patient's own perception of spasms, spasticity-related pain and other associated symptoms is not common in daily neurological practice. Some of the usual spasticity scales, such as the Ashworth and modified Ashworth scales, reflect the observer's measurement of spasticity at a particular point in time. Herbal (smoked) cannabis has long been recognized as a possible option for relief of spasticity and neuropathic pain, but pertinent concerns about psychoactive effects and addiction risk have prevented its common use. An innovative method of benefiting from the mode of action of cannabinoids while limiting their drawbacks is to reduce peak plasma levels of 9-delta-tetrahydrocannabinol and counteract psychoactivity with higher than naturally occurring proportions of a second cannabinoid, cannabidiol. Sativex® oromucosal spray (1:1 ratio of 9-delta-tetrahydrocannabinol/cannabidiol) has recently been approved in a number of EU countries and elsewhere for use in patients with MS-related spasticity who are resistant to treatment with other antispasticity medications. In clinical trials, Sativex provided initial relief of spasticity symptoms within the first 4 weeks of treatment (trial period) in up to about half of patients resistant to other available oral antispasticity medications and demonstrated clinically significant improvement in spasticity (30% or higher reduction from baseline) in three-quarters of the initial responders. Adverse events were limited mainly to mild or moderate cases of somnolence and dizziness. Under everyday clinical practice conditions, Sativex at a mean daily dose of <7 sprays/day, was shown to relieve spasticity in about 70% of patients previously resistant to treatment. Clear improvements were also noted in associated symptoms such as sleep disturbances, bladder problems, loss of mobility and cramps. In large observational studies, >80% of patients reported no adverse events with the use of Sativex and interim data from safety registries in the UK and Spain indicate a low risk for serious adverse drug reactions. Follow-up studies in Sativex responders support continued benefit without the need to increase doses for at least 1 year. Sativex appears to be a promising solution for a meaningful proportion of patients with MS-related spasticity who have inadequate response to current antispasticity medications.
Article
To gain real-life data on demographic and clinical characteristics, treatment patterns, treatment satisfaction and quality-of-life of multiple sclerosis-related spasticity (MSS) in Germany. MObility ImproVEment (MOVE 1), a cross-sectional burden-of-disease study, combines retrospective 12-month chart documentation with questionnaires for both, patients and physicians. Data were collected at office-based neurologists, MS outpatient clinics and rehabilitation centres in Germany. Structured documentation forms, questionnaires and validated instruments were used for data collection. Patients with mild to severe MSS were included. Participants documented the clinical characteristics, impact of MSS on daily living, quality-of-life, treatment patterns and satisfaction with available drug treatment stratified by severity of MSS. Severity was assessed by patients and physicians. Of 419 patients enrolled at 42 centres from 4/2011 to 9/2011, 414 were available for analysis (mean age: 48.5 years; female: 64%). Most disturbing symptoms associated with spasticity reported by physicians and patients were stiffness (74%) and mobility restrictions (66%). Mean EQ-5D score fell from 0.6 to 0.3 with increasing severity of spasticity, while percentage of subjects with spasticity-related impairment of activities every day rose from 10% in patients with mild to 85% in patients with severe spasticity. At time of enrolment, 55% of patients received pharmacotherapy and 78% physiotherapy. These percentages increased with increasing severity (drugs: 39-84%; physiotherapy: 65-86%). Overall, 41% of physicians and 36% of patients were partial dissatisfied or dissatisfied with the effectiveness of available anti-spastic pharmacotherapy. Spasticity and its symptoms impair personal well-being and quality-of-life. Treatment of spasticity with drugs and physiotherapy is common, but satisfaction with the currently available anti-spastic pharmacotherapy is low.