ArticlePDF AvailableLiterature Review

Pharmacotherapeutic considerations for use of cannabinoids to relieve pain in patients with malignant diseases

Taylor & Francis
Journal of Pain Research
Authors:

Abstract and Figures

Purpose The aim of this review was to assess the efficacy of cannabis preparations for relieving pain in patients with malignant diseases, through a systematic review of randomized controlled trials (RCTs), which were predominantly double-blind trials that compared cannabis preparation to a placebo. Methods An electronic search of all literature published until June 2017 was made in MEDLINE/PubMed, Embase, The Cochrane Controlled Trials Register and specific web pages devoted to cannabis. Results Fifteen of the 18 trials demonstrated a significant analgesic effect of cannabinoids as compared to placebo. The most commonly reported adverse effects were generally well tolerated, mild to moderate. The main side effects were drowsiness, nausea, vomiting and dry mouth. There is evidence that cannabinoids are safe and modestly effective in neuropathic pain and also for relieving pain in patients with malignant diseases. The proportion of “responders” (patients who at the end of 2 weeks of treatment reported ≥30% reduction in pain intensity on a scale of 0–10, which is considered to be clinically important) was 43% in comparison with placebo (21%). Conclusion The target dose for relieving pain in patients with malignant diseases is most likely about 10 actuations per day, which is about 27 mg tetrahydrocannabinol (THC) and 25 mg cannabidiol (CBD), and the highest approved recommended dose is 12 actuations per day (32 mg THC/30 mg CBD). Further large studies of cannabinoids in homogeneous populations are required.
This content is subject to copyright. Terms and conditions apply.
© 2018 Darkovska-Serafimovska et al. This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.
php and incorporate the Creative Commons Attribution – Non Commercial (unported, v3.0) License (http://creativecommons.org/licenses/by-nc/3.0/). By accessing the work
you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For
permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms (https://www.dovepress.com/terms.php).
Journal of Pain Research 2018:11 837–842
Journal of Pain Research Dovepress
submit your manuscript | www.dovepress.com
Dovepress 837
REVIEW
open access to scientific and medical research
Open Access Full Text Article
http://dx.doi.org/10.2147/JPR.S160556
Pharmacotherapeutic considerations for use
of cannabinoids to relieve pain in patients with
malignant diseases
Marija
Darkovska-Serafimovska1
Tijana Serafimovska2
Zorica Arsova-Sarafinovska1
Sasho Stefanoski3
Zlatko Keskovski3
Trajan Balkanov4
1Department of Pharmacology,
Faculty of Medical Sciences, Goce
Delcev University, Stip, Republic of
Macedonia; 2Faculty of Pharmacy, Ss
Cyril and Methodius University of
Skopje, Skopje, Republic of Macedonia;
3NYSK Holdings, Skopje, Republic
of Macedonia; 4Department of
Pharmacology and Toxicology, Faculty
of Medicine, Ss Cyril and Methodius
University of Skopje, Skopje, Republic
of Macedonia
Purpose: The aim of this review was to assess the efficacy of cannabis preparations for
relieving pain in patients with malignant diseases, through a systematic review of randomized
controlled trials (RCTs), which were predominantly double-blind trials that compared cannabis
preparation to a placebo.
Methods: An electronic search of all literature published until June 2017 was made in MED-
LINE/PubMed, Embase, The Cochrane Controlled Trials Register and specific web pages
devoted to cannabis.
Results: Fifteen of the 18 trials demonstrated a significant analgesic effect of cannabinoids as
compared to placebo. The most commonly reported adverse effects were generally well tolerated,
mild to moderate. The main side effects were drowsiness, nausea, vomiting and dry mouth. There
is evidence that cannabinoids are safe and modestly effective in neuropathic pain and also for
relieving pain in patients with malignant diseases. The proportion of “responders” (patients who
at the end of 2 weeks of treatment reported 30% reduction in pain intensity on a scale of 0–10,
which is considered to be clinically important) was 43% in comparison with placebo (21%).
Conclusion: The target dose for relieving pain in patients with malignant diseases is most likely
about 10 actuations per day, which is about 27 mg tetrahydrocannabinol (THC) and 25 mg can-
nabidiol (CBD), and the highest approved recommended dose is 12 actuations per day (32 mg
THC/30 mg CBD). Further large studies of cannabinoids in homogeneous populations are required.
Keywords: cancer management, chronic pain, cannabidiol, tetrahydrocannabinol, medical
marihuana, nabiximols, cannabinoid receptors
Introduction
Pain is a disagreeable sensorial and emotional experience that subjects associate with
tissue damage and impairs quality of life.1 Effective therapeutic options for patients
living with different forms of pain are limited. Opioids and anti-inflammatory drugs
as first-line medications for the treatment of pain in patients with malignant diseases
do not always give satisfactory results.
In traditional medicine, cannabis preparations have been used for thousands of
years to treat disease or alleviate symptoms, but their efficacy for specific indications
is not clear. Clinical use of cannabinoid substances is restricted, due to legal and ethical
reasons, as well as limited evidence showing benefits. The medical use of cannabis is
attractive to patients suffering from malignant diseases. However, scientific justifica-
tion or positive experience of the use of cannabis in patients with malignant diseases
has only been found for the following indications: alleviation of sickness, nausea and
Correspondence: Marija Darkovska-
Seramovska
Department of Pharmacology, Faculty
of Medical Sciences, Goce Delcev
University, “Krste Misirkov” 10-A. PO
201, Stip, 2000, Republic of Macedonia
Email marija.darkovska@udg.edu.mk
Journal name: Journal of Pain Research
Article Designation: REVIEW
Year: 2018
Volume: 11
Running head verso: Darkovska-Serafimovska et al
Running head recto: Cannabinoids for relief of pain in patients with malignant diseases
DOI: http://dx.doi.org/10.2147/JPR.S160556
This article was published in the following Dove Press journal:
Journal of Pain Research
Journal of Pain Research 2018:11
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
838
Darkovska-Seramovska et al
vomiting associated with the use of cytotoxic therapy, pain
relief and stimulation of appetite (treatment of cachexia).
Pain relief is the most commonly cited reason for the medi-
cal use of cannabis.2–5
Mechanism of action of cannabinoids
Cannabinoids bind to cannabinoid receptors and act as ago-
nists. Cannabinoid receptors are cell membrane receptors,
members of the G protein-coupled receptors. They are acti-
vated by three major groups of ligands: endocannabinoids,
plant cannabinoids and synthetic cannabinoids. Four subtypes
of these receptors have been identified. Two have been cloned
(CB1 and CB2 cannabinoid receptors),6,7 while the other two,
WIN and abnormal-cannabidiol (abn-CBD) receptors, have
been characterized pharmacologically.8–11
The analgesic effect of cannabinoids as a result of binding
of cannabinoids to cannabinoid receptors has been confirmed,
and the role of the endocannabinoid system in pain relief
has been verified in various types of pain: somatic, visceral
and neuropathy.12 Classical analgesics, nonsteroidal anti-
inflammatory drugs or opioids, paracetamol and antidepres-
sants (with an analytical effect in some conditions) increase
the activity of the endocannabinoid system.12
The discovery of the endocannabinoid system and the
development of animal models with different forms of pain
have recently demonstrated the synergism between the opi-
ate and cannabinoid systems.12 There is a large amount of
preclinical data in animal models on the analgesic effect
of cannabinoids, predominantly Δ9-tetrahydrocannabinol
(THC), nabilone and dronabinol, or combinations of THC and
cannabidiol (CBD) and some other synthetic cannabinoids;
and analgesic effects in the treatment of cancer-related pain
without serious side effects have been shown.13–15
In humans, pharmacodynamic studies have demonstrated
the effect of cannabinoids on provoked somatic pain (e.g.,
thermal stimulation), capsaicin-induced hyperalgesia, painful
spasms in patients with multiple sclerosis (MS), and neuro-
pathic pain in HIV/AIDS patients.12,26–28,30
Available cannabinoid analgesic agents
Two finished drug products – nabilone (Cesamet) and
dronabinol (Marinol) – have been approved in many coun-
tries for the “prevention/treatment of chemotherapy induced
nausea and vomiting”.15 The only pharmaceutical industry
drug product carrying the cannabinoid therapeutic principle
with regulatory approval (in some countries) for pain relief
in patients with malignant diseases is nabiximols (Sativex
spray). Sativex® (GW Pharmaceuticals, Cambridge, UK)
is an oromucosal cannabis-based spray combining a CB1
partial agonist (THC) with a cannabinoid system modulator
(CBD).16,17 It was approved by Health Canada in June 2005
for prescription for central neuropathic pain in MS, and in
August 2007, it was additionally approved for the treatment of
cancer pain, as an adjuvant analgesic in adults with advanced
malignancy, who, despite the highest tolerated opioid dose,
still feel moderate to severe chronic pain.18
In randomized controlled trials (RCTs), Sativex was
adjunctively added to optimal drug regimens in patients with
intractable symptoms, those often termed “untreatable”. The
recommended maximum is 12 daily doses (32.4 mg THC and
30 mg CBD).12 Data on the analgesic effect of nabiximols in
malignant patients are shown primarily as an illustration of
the effects of different ratios of THC/CBD.
Approved ongoing clinical trials
An investigational new drug (IND) application to study
Sativex in advanced clinical trials in the USA was approved
by the FDA in January 2006 in patients with intractable
cancer pain.15
Recently, the European Medicine Agency (EMEA)
approved two double-blind, placebo-controlled safety and
efficacy studies of Sativex as adjunctive therapy to opiates:
the first one in pediatric patients from 8 to less than 18
years of age with cancer-related pain and the second one
in pediatric patients from birth to less than 8 years of age
with cancer-related pain (decision number P/0298/2014, PIP
number EMEA-000181-PIP02-13). The completion date of
the pediatric investigation plan is by July 2026.19
On February 17, 2016, orphan designation (EU/3/16/1621)
was granted by the European Commission to GW Pharma-
ceuticals, for THC and CBD from extracts of Cannabis sativa
for the treatment of glioma.20
Methods
An electronic search of all literature published until June 2017
was made in MEDLINE/PubMed, Embase, The Cochrane
Controlled Trials Register and specific web pages devoted to
cannabis. A systematic review of literature identified RCTs,
evaluating the efficacy of cannabinoids in various chronic
pain conditions that are not related to malignant diseases
(including MS and HIV/AIDS neuropathies), compared with
placebo and sometimes other active treatments.21–38 They
demonstrated an analgesic effect of dronabinol, nabilone
and natural THC and CBD in comparison with smoking
marijuana. A detailed overview of preclinical and clinical
data on the analgesic efficacy of cannabinoids is found in the
Journal of Pain Research 2018:11 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
839
Cannabinoids for relief of pain in patients with malignant diseases
document Health Canada2 and Ethnopharmacology.39 Only
three RTCs evaluated the efficacy of cannabinoids compard
to placebo in various pain conditions that are related to
malignant diseases.40-42 The studies selected were double-
blind RTCs with a crossover or parallel design.
Selection criteria
A systematic review of literature using the keywords canna-
binoids, pain, malignant diseases, THC and RCTs identified
198 reports, of which 73 were potentially relevant RCTs,
based on title and abstract screening. Seven of them had
no relevant information obtained as full-text studies, five
reports were duplicates (they contained data that had previ-
ously been published), 60 were in other clinical examinations
(chemotherapy-induced nausea and vomiting, spasticity due
to MS, sleep disorders and HIV/AIDS) and ten compared
efficacy of cannabis for the treatment of chronic pain. Only
three RCTs evaluated the efficacy of cannabinoids in pain
conditions that are related to malignant diseases compared
with synthetic THC and placebo (Figure 1).40–42
Review of relevant research
In the intervention group, subjects were required to
have received cannabis preparation, which at minimum
contained the cannabinoids THC and CBD, applied by
oral administration. Synthetic derivates of THC, such as
dronabinol, nabilone or benzopyranoperidine, were likewise
included. In the control group, subjects were required to have
received a placebo treatment.40–42
The measure of efficacy chosen was the variable “inten-
sity of pain” as scored by numeric analog scales. Patients at
the end of 2 weeks of treatment reported 30% reduction in
pain intensity on a scale of 0–10, which was considered as
clinically important.
Double-blind, 2-week, multicenter RCT,
placebo-controlled study
Respondents to the double-blind, 2-week, multicenter RCT,
placebo-controlled study40 were adult patients with malignant
diseases in terminal stage, who for at least a week used high
(the most tolerated) doses of strong opiates, and despite this
showed an intensity of pain 4 on a scale of 0–10.
For the treatment, patients were randomized into three
groups. The first group received Sativex oral spray (nabixi-
mols, 2.7 mg THC, 2.5 mg CBD per actuation) (n=60), the
second group received THC as oral spray (2.7 mg THC per
actuation) (n=58) and the third group received placebo spray
(n=59). During the first week, the dose was titrated in patients
based on tolerability and analgesia. The maximum permis-
sible dose was 8 actuations in 3 hours (in intervals of at least
15 minutes between two doses) or at most 48 actuations for
24 hours (130 mg THC and 120 mg CBD).
The proportion of “responders” (patients who at the end
of 2 weeks of treatment reported 30% reduction in pain
intensity on a scale of 0–10, which is considered to be clini-
cally important) in the Sativex group was 43% (statistically
significantly compared with placebo; an improvement of
–1.37 vs –0.69); in the THC group 23% (non-significant
change compared with placebo; an improvement of –1.01
vs –0.69) and in the placebo group 21%.
Regarding side effects, in the Sativex group, 10/60
patients dropped out of treatment because of the side effects;
in the second group where THC was orally administrated as
spray, 7/58 patients dropped out of treatment because of the
side effects and in the placebo group, 3/59 patients dropped
out of treatment because of the side effects. The main side
effects were drowsiness, nausea, vomiting and dry mouth.
Extension of the main double-blind,
2-week, multicenter RCT, placebo-
controlled study
In 2013, an extension of the main study was opened.28 A total
of 39 patients with chronic pain due to malignant disease,
198 Title and abstracts screened
125 Excluded reports
73 Full reports assessed
70 Excluded reports:
28 Chemotherapy-induced nausea
and vomiting
14 Spasticity due to multiple sclerosis
10 Chronic pain
7 No relevant information
5 Duplicates
4 HIV/AIDS
2 Sleep disorders
3 RCTs included – evaluating the
efficacy of cannabinoids in pain
conditions that are related to
malignant diseases
Figure 1 Flow of studies through the review process.
Abbreviation: RCTs, randomized controlled trials.
Journal of Pain Research 2018:11
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
840
Darkovska-Seramovska et al
who had been treated with opioids in a previous study but
with an inadequate analgesic response, were involved in a
new extended open, randomized, controlled, multicenter
study for 2 weeks in which patients continued treatment with
Sativex.41 Doses remained unchanged. Of the 39 patients, 15
were treated for less than 2 weeks; others gradually withdrew
from treatment during 1 year due to side effects (23/39), loss
of efficacy (3/39) and a number of other reasons.
The study showed that the long-term use of THC/CBD
spray is generally well tolerated without any loss of effect
for pain relief because of long-term use. Moreover, patients
who continued to use spray did not seek to increase the dose
of spray or other medications for pain relief, suggesting
that the adjuvant use of cannabinoids in cancer-related pain
could be useful.
Multicenter, double-blind, placebo-
controlled RCT, three different doses of
Sativex
Respondents to this multicenter, double-blind, placebo-
controlled RCT, with three different doses of Sativex,42 were
adult patients with malignant diseases in terminal stage, who
for at least a week used high (the most tolerated) doses of
strong opiates, and despite this showed an intensity of pain
4 on a scale of 0–10.
For treatment, patients were randomized to receive three
different doses of Sativex: group 1 received 4 actuations per
day (10.8 mg THC, 10 mg CBD) (n=91), group 2 received
10 actuations per day (27 mg THC, 25 mg CBD) (n=88)
and group 3 received 16 actuations per day (43 mg THC, 40
mg CBD) (n=90). A group receiving THC as oral spray and
placebo spray (n=90) was also included. During the first 7
days, the dose was gradually increased (from 1 to 4, 10 or
16) and then adjusted for the next 2 weeks. From the 21st to
the 35th day, there was a 14-day evaluation period.
Patients who at the end of 2 weeks of treatment reported
30% reduction in pain intensity on a scale of 0–10 were
considered clinically important.
The results showed that with increasing doses, several
patients dropped out of treatment because of side effects:
3/91 placebo, 5/91 lowest dose, 6/88 medium dose and 20/90
highest dose. Overall, the share of those with 30% reduction
in pain did not differ for Sativex vs placebo. However, in an
analysis that evaluated the average daily pain score during 14
days, the lowest and midpoint doses were better than placebo.
There is no RTC for relief of pain in malignant diseases
that evaluates “smoking marijuana” or some other herbal prep-
aration consisting of natural THC and CBD, except Sativex.
Discussion
The natural and synthetic agonists of the cannabinoid recep-
tors have shown positive therapeutic results in the treatment
of various pathological conditions, including pain as an
inevitable symptom of tissue damage. The antinociceptive
and anti-hyperalgesic effect of cannabinoids at the peripheral
and central levels has been demonstrated and confirmed in
various models of acute and chronic pain.43,44
The adverse effects (AEs) of cannabinoids on the cen-
tral nervous system (CNS) are associated with abnormal
psychomotor behavior, short-term memory impairment and
intoxication.45
This review is prepared according to recommendations for
systematic reviews.46,47
A systemic literature review identified
more RCTs evaluating the efficacy of cannabinoids/cannabis
in pain conditions, but only three double-blind controlled
RTCs comparing the effectiveness of cannabinoids with
synthetic THC and placebo in a variety of painful conditions
that are associated with malignant disease.
In the first double-blind, 2-week multicenter RCT, pla-
cebo-controlled study,40 the proportion of responders in the
Sativex group was 43%, in the THC group was 23% and in
the placebo group was 21%. Extension of the main double-
blind, 2-week, multicenter RCT, placebo-controlled study41
in 2013 showed that long-term use of THC/CBD spray is
generally well tolerated without any loss of effect for pain
relief because of long-term use. A multicenter double-blind,
placebo-controlled RCT, in which three different doses
of Sativex were used,42 showed that with increasing doses
of THC/CBD, several patients dropped out of treatment
because of side effects and those with 30% reduction in
pain did not differ for Sativex vs placebo.
Thus, in two high-quality studies, the primary outcome
of one can be regarded as positive and the other negative;
however, both can be considered as evidence of the effective-
ness of Sativex in specific painful conditions.
The inefficacy of the THC oral spray in one study, and
variable results with relatively high doses of dronabinol,
indicate that THC alone may not be sufficient for a good
analgesic effect. This means that THC should be combined
with CBD in order to achieve desire results.
Medicines that contain active ingredients that act as ago-
nists of cannabinoid receptors are a promising therapeutic
approach for treatment of various types of pain: neuropathic,
inflammatory and oncological. This primarily refers to prepa-
rations containing exactly known similar amounts of THC
and CBD, intended for the treatment of patients who do not
respond to conventional therapy.11,41,42
Journal of Pain Research 2018:11 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
841
Cannabinoids for relief of pain in patients with malignant diseases
In the studies of Sativex in patients with malignant dis-
eases and pain despite opioid therapy, the target dose is most
likely about 10 actuations per day (the average number of
actuations in one RCT was 9; in the second, the “successful”
dose was 6–10 actuations per day), which is about 27 mg THC
and 25 mg CBD, and the highest approved recommended dose
is 12 actuations per day (32 mg THC/30 mg CBD). The dose
is introduced gradually, from 1 to 4–6 actuations during the
day, for 5–7 days.12,37
Conclusion
There is evidence, although limited, to support the use of can-
nabis pharmacotherapy in the treatment of different forms of
pain in patients. If a patient with chronic pain and their health-
care provider work together through first- and second-line
treatment modalities without success, a trial of cannabis or
a cannabinoid may be a reasonable next step. With increased
use of medical cannabis as pharmacotherapy for pain comes
a need for comprehensive risk–benefit discussions that take
into account the significant possible AEs of cannabis.
Numerous randomized clinical trials have demonstrated
the safety and efficacy of Sativex in central and peripheral
neuropathic pain, rheumatoid arthritis and cancer pain. Com-
mon AEs included dizziness, dry mouth, nausea, fatigue,
somnolence, euphoria and vomiting.
The recommended daily dose for the treatment of pain
is a maximum of 32.4 mg THC and 30 mg CBD. Data on
the analgesic effect of nabiximols in patients with malignant
diseases are shown primarily as an illustration of the effects
of different ratios of the THC/CBD combination. The degree
to which cannabinoid analgesics will be adopted in adjunc-
tive pain management practices remains to be determined.
Further large studies of cannabinoids in homogeneous
populations are required.
Author contributions
All authors contributed toward data analysis, drafting and
critically revising the paper and agreed to be accountable
for all aspects of the work.
Disclosure
The authors report no conflicts of interest in this work.
References
1. Classification of Chronic Pain. Descriptions of chronic pain syndromes
and definitions of pain terms. Prepared by the International Associa-
tion for the Study of Pain, Subcommittee on Taxonomy. Pain Suppl.
1986;3:S1–226. Available from: https://s3.amazonaws.com/rdcms-iasp/
files/production/public/Content/ContentFolders/Publications2/Free-
Books/Classification-of-Chronic-Pain.pdf. Accessed March, 24, 2018.
2. Bestrashniy J, Winters KC. Variability in medical marijuana laws in the
United States. Psychol Addict Behav. 2015;29:639–642.
3. Light MK, Orens A, Lewandowski B, et al. Market size and demand for
marijuana in Colorado. The Marijuana Policy Group. 2014. Available
from: http://www.cannabisconsumer.org/uploads/9/7/9/6/97962014/
market_size_and_demand_study_july_9_2014%5B1%5D.pdf.
Accessed November 17, 2016.
4. Ilgen MA, Bohnert K, Kleinberg F, et al. Characteristics of adults seeking
medical marijuana certification. Drug Alcohol Depen. 2013;132:654–659.
5. Hill KP, Palastro MD, Johnson B, Ditre JW. Cannabis and pain: a clinical
review. Cannabis Cannabinoid Res. 2017;2(1):96–104.
6. Matsuda LA, Lolait SJ, Brownstein MJ, Young AC, Bonner TI. Structure
of a cannabinoid receptor and functional expression of the cloned cDNA.
Nature. 1990;346:561–564.
7. Munro S, Thomas KL, Abu-Shaar M. Molecular characterization of a
peripheral receptor for cannabinoids. Nature. 1993;365:61–65.
8. Breivogel CS, Griffin G, Di Marzo V, Martin BR. Evidence for a new
G-protein-coupled cannabinoid receptor in mouse brain. Mol Pharma-
col. 2001;60:155–163.
9. Di Marzo V, Breivogel CS, Tao Q, et al. Levels, metabolism, and pharma-
cological activity of anandamide in CB1 cannabinoid receptor knockout
mice, evidence for non-CB1, non-CB2 receptor-mediated actions of
anandamide in mouse brain. J Neurochem. 2000;75:2434–2444.
10. Hájos N, Ledent C, Freund TF. Novel cannabinoid sensitive receptor
mediates inhibition of glutamatergic synaptic transmission in the hip-
pocampus. Neuroscience. 2001;106:1–4.
11. Manzanares J, Julian MD, Carrascosa A. Role of the cannabinoid system
in pain control and therapeutic implications for the management of acute
and chronic pain episodes. Curr Neuropharmacol. 2006;4(3):239–257.
12. Health Canada [webpage on the Internet]. Information for health care pro-
fessionals. Cannabis (marihuana, marijuana) and the cannabinoids. Health
Canada; 2013. Available from: https://www.canada.ca/en/health-canada/
services/drugs-health-products/medical-use-marijuana/information-
medical-practitioners/information-health-care-professionals-cannabis-
marihuana-marijuana-cannabinoids.html. Accessed March, 24, 2018.
13. Cridge BJ, Rosengren RJ. Critical appraisal of the potential use of cannabi-
noids in cancer management. Cancer Management Res. 2013;5:301–315.
14. McAllister SD, Soroceanu L, Desperz PY. The antitumor activity of
plant-derived non-psychoactive cannabinoids. J Neuroimmune Phar-
macol. 2015;10(2):255–267.
15. Abrams DI, Guzman M. Cannabis in cancer care. Clin Pharmacol Ther.
2015;97:575–586.
16. McPartland JM, Russo EB. Cannabis and cannabis extracts: greater
than the sum of their parts? J Cannabis Therapeut. 2001;1:103–132.
17. Russo EB, Guy GW. A tale of two cannabinoids: the therapeutic rationale
for combining tetrahydrocannabinol and cannabidiol. Med Hypotheses.
2006;66:234–46.
18. Russo EB. Cannabinoids in the management of difficult to treat pain.
Ther Clin Risk Manag. 2008;4(1):245–259.
19. European Medicines Agency [webpage on the Internet]. EMEA-001902-
PIP01-15-M01. Available from: http://www.ema.europa.eu/ema/index.
jsp?curl=pages/medicines/pips/EMEA-000181-PIP02-13/pip_001257.
jsp&mid=WC0b01ac058001d129. Accessed March, 24, 2018.
20. European Medicines Agency [webpage on the Internet]. EU/3/16/1621.
Available from: http://www.ema.europa.eu/ema/index.jsp?curl=pages/
medicines/human/orphans/2016/04/human_orphan_001722.
jsp&mid=WC0b01ac058001d12b. Accessed March, 24, 2018.
21. Wade DT, Collin C, Stott C, Duncombe P. Meta-analysis of the efficacy
and safety of Sativex (nabiximols) on spasticity in people with multiple
sclerosis. Mult Scler. 2010;16:707–714.
22. Zajicek J, Ball S, Wright D, et al. Effect of dronabinol on progression
in progressive multiple sclerosis (CUPID): a randomized, placebo-
controlled trial. Lancet Neurol. 2013;12:857–865.
23. Ball S, Vickery J, Hobart J, et al. The cannabinoid use in progressive
inflammatory brain disese (CUPID) trial: a randomized dobule-blind
placebo-controlled parallel-group multicenter trial and economic evalu-
ation of cannabinoids to slow progression in multiple sclerosis. Health
Technol Assess. 2015;19(12):vii–viii, xxv–xxxi, 1–187.
Journal of Pain Research 2018:11
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
Journal of Pain Research
Publish your work in this journal
Submit your manuscript here: https://www.dovepress.com/journal-of-pain-research-journal
The Journal of Pain Research is an international, peer reviewed, open
access, online journal that welcomes laboratory and clinical findings
in the fields of pain research and the prevention and management
of pain. Original research, reviews, symposium reports, hypoth-
esis formation and commentaries are all considered for publication.
The manuscript management system is completely online and includes
a very quick and fair peer-review system, which is all easy to use. Visit
http://www.dovepress.com/testimonials.php to read real quotes from
published authors.
Dovepress
842
Darkovska-Seramovska et al
24. Collin C, Davies P, Mutiboko IK, Ratcliffe S; Sativex Spasticity in MS
Study Group. Randomized controlled trial of cannabis-based medicine
in spasticity caused by multiple sclerosis. Eur J Neurol. 2007;14:
290–296.
25. Collin C, Ehler E, Waberzinek G, et al. A double-blind, random-
ized, placebo-controlled, parallel group study of Sativex in subjects
with symptoms of spasticity due to multiple sclerosis. Neurol Res.
2010;32:451–459.
26. Notcutt W, Langford R, Davies P, Ratcliffe S, Potts R. A placebo-
controlled, parallel-group randomized withdrawal study of subjects
with symptoms of spasticity due to multiple sclerosis who are receiving
long-term Sativex (nabiximols). Mult Scler. 2012;18:219–228.
27. 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;18:1122–1131.
28. Wade DT, Makela P, Robson P, House H, Bateman C. Do cannabis-
based medicinal extracts have general or specific effects on symptoms
in multiple sclerosis? A double-blind, randomized, placebo-controlled
study of 160 patients. Mult Scler. 2004;10:434–441.
29. Zajicek J, Fox P, Sanders H, et al. Cannabinoids for treatment of
spasticity and other symptoms related to multiple sclerosis (CAMS
study): multicenter randomized placebo-controlled trial. Lancet.
2003;362:1517–1526.
30. Corey-Bloom J, Wolfson T, Gamst A, et al. Smoked cannabis for spastic-
ity in multiple sclerosis: a randomized, placebo-controlled trial. CMAJ.
2012;184:1143–1150.
31. Rog DJ, Nurmikko TJ, Friede T, Young CA. Randomized controlled
trial of cannabis-based medicine in central pain in multiple sclerosis.
Neurology. 2005;65:812–819.
32. Phillips TJC, Cherry CL, Cox S, Marshall SJ, Rice AS. Pharmacological
treatment of painful HIV-associated sensory neuropathy: a systematic
review and meta-analysis of randomized controlled trials. Plos One.
2010;5(12):e14433.
33. Abrams DI, Hilton JF, Leiser RJ, et al. Short-term effects of cannabi-
noids in patients with HIV-1 infection: a randomized placebo-controlled
clinical trial. Ann Intern Med. 2003;139:258–266.
34. Abrams DI, Jay CA, Shade SB, et al. Cannabis in painful HIV-associated
sensory neuropathy. Neurology. 2007;68:515–521.
35. Ellis RJ, Toperoff W, Vaida F, van den Brande G, et al. Smoked medicinal
cannabis for neuropathic pain in HIV: a randomized, crossover clinical
trial. Neuropsychopharmacology. 2009;34:672–680.
36. Ashton JC. Emerging treatment options for spasticity in multiple sclerosis –
clinical utility of cannabinoids. Deg Neurol Neuromusc Dis. 2011;1:15–23.
37. Lynch ME, Campbell F. Cannabinoids for treatment of chronic non-
cancer pain; a systematic review of randomized trials. Br J Clin Phar-
macol. 2011;72:735–744.
38. Martín-Sánchez E, Fur ukawa TA, Taylor J, Martin JL. Systematic review
and meta-analysis of cannabis treatment for chronic pain. Pain Med.
2009;10:1353–1368.
39. Ben Amar M. Cannabinoids in medicine: a review of their therapeutic
potential. J Ethnopharmacol. 2006;105:1–25.
40. Johnson JR, Burnell-Nugent M, Lossignol D, Ganae-Motan ED, Potts R,
Fallon MT. Multicenter, double-blind, randomized, placebo-controlled,
parallel-group study of the efficacy, safety and tolerability of THC:CBD
extract and THC extract in patients with intractable cancer-related pain.
J Pain Symptom Manage. 2010;39:167–179.
41. Johnson JR, Lossignol D, Burnell-Nugent M, Fallon MT. An open-label
extension study to investigate the long-term safety and tolerability of
THC/CBD oromucosal spray and oromucosal THC spray in patients
with terminal cancer-related pain refractory to strong opioid analgesics.
J Pain Symptom Manage. 2013;46:207–218.
42. Portenoy RK, Banae-Motan ED, Allende S, et al. Nabiximols for opioid-
treated cancer patients with poorly-controlled chronic pain: a random-
ized placebo-controlled dose-graded trial. J Pain. 2012;13:438–449.
43. Iversen L, Chapman V. Cannabinoids, a real prospect for pain relief?
Curr Opin Pharmacol. 2002;2:50–55.
44. Pertwee RG. Cannabinoid receptors and pain. Prog Neurobiol.
2001;63:569–611.
45. Iversen L. Cannabis and the brain. Brain. 2003;126:1252–1270.
46. Higgins JPT, Green S, eds. Cochrane Handbook for Systematic Reviews
of Interventions [webpage on the Internet]. Version 5.1.O (updated
March 2011). Available from: http://handbook.cochrane.org/. Accessed
June 23, 2017.
47. Centre for Reviews and Dissemination. Systematic Reviews: CRD’s
Guidance for Undertaking Reviews in Health Care. York, UK: Univer-
sity of York; 2009. Available from: https://www.york.ac.uk/media/crd/
Systematic_Reviews.pdf. Accessed June 23, 2017.
... Eight systematic reviews claimed safety based on between 386 and 3056 patients and 0 to 325 serious adverse events 13,21,34,[37][38][39]58,60 (Supplementary file 2, available at http://links. lww.com/PAIN/B967). ...
... Eleven of the 65 systematic reviews with abstracts (17%) were subject to moderate or severe narrative bias ( Table 4; scoring for all systematic reviews is in Supplementary file 4, available at http://links.lww.com/PAIN/B967) 12,13,21,34,[37][38][39]58,60,63,65 : 6 reviews because of inappropriate claims of safety only, 21,34,37,38,58,60 3 for inappropriate claims of efficacy alone, 12,63,65 and 2 for inappropriate claims of safety and efficacy. 13,39 Claims of safety were based on 386 to 3056 patients with proportions of serious adverse events up to 11% or a 95% confidence in the upper limit of serious adverse events of 1 in 451. ...
... 12,13,21,34,[37][38][39]58,60,63,65 : 6 reviews because of inappropriate claims of safety only, 21,34,37,38,58,60 3 for inappropriate claims of efficacy alone, 12,63,65 and 2 for inappropriate claims of safety and efficacy. 13,39 Claims of safety were based on 386 to 3056 patients with proportions of serious adverse events up to 11% or a 95% confidence in the upper limit of serious adverse events of 1 in 451. ...
Article
We define narrative bias as a tendency to interpret information as part of a larger story or pattern, regardless of whether the facts support the full narrative. Narrative bias in title and abstract means that results reported in the title and abstract of an article are done so in a way that could distort their interpretation and mislead readers who had not read the whole article. Narrative bias is often referred to as “spin.” It is prevalent in abstracts of scientific papers and is impactful because abstracts are often the only part of an article read. We found no extant narrative bias instrument suitable for exploring both efficacy and safety statements in randomized trials and systematic reviews of pain. We constructed a 6-point instrument with clear instructions and tested it on randomised trials and systematic reviews of cannabinoids and cannabis-based medicines for pain, with updated searches to April 2021. The instrument detected moderate or severe narrative bias in the title and abstract of 24% (8 of 34) of randomised controlled trials and 17% (11 of 64) of systematic reviews; narrative bias for efficacy and safety occurred equally. There was no significant or meaningful association between narrative bias and study characteristics in correlation or cluster analyses. Bias was always in favour of the experimental cannabinoid or cannabis-based medicine. Put simply, reading title and abstract only could give an incorrect impression of efficacy or safety in about 1 in 5 papers reporting on these products.
... [39] Chronic pain and multiple sclerosis Opioids and anti-inflammatory drugs are usually the first line treatment toward pain associated with malignant diseases. Sometimes they do not achieve satisfactory results [40] Pharmacodynamic studies have demonstrated the effect of cannabinoids on capsaicin-induced hyperalgesia, neuropathic pain in HIV/AIDS patients, painful spasms in patients with multiple sclerosis-provoked somatic pain (e.g., thermal stimulation). [40] Dronabinol and Nabilone have shown efficacy in the treatment of pain and distress. ...
... Sometimes they do not achieve satisfactory results [40] Pharmacodynamic studies have demonstrated the effect of cannabinoids on capsaicin-induced hyperalgesia, neuropathic pain in HIV/AIDS patients, painful spasms in patients with multiple sclerosis-provoked somatic pain (e.g., thermal stimulation). [40] Dronabinol and Nabilone have shown efficacy in the treatment of pain and distress. [10] Although cannabis plant has not been approved for medical use, several drugs that contain individual cannabinoids have been approved by FDA as shown in Table 1. ...
Article
Full-text available
The use of cannabis for medical purposes has been a subject for discussion for so many years. Cannabis as a source of medical treatment first came to light in the 19th century. However, origins of cultivation of marijuana as a medical plant can be traced back to thousands of years. Attempts to completely legalize the use of cannabis for medical purposes are strongly contested in many places due to some of its negative effects on users physically, psychologically, and socially. This review is aimed to discuss the mechanism of action and pharmacogenetics of cannabinoids to elucidate its uses as medicine as well as negative effects. Online searches on the following database: Google Scholar, PubMed, Biomed Central, and SciELO were done. An attempt was made to review articles with keywords such as cannabis, cannabinoid receptors, genes, and medical marijuana. This review has highlighted with evidence the importance of genomic profiling to prevent side effects associated with predisposing genes for the benefit of patients who are medical candidates for medicinal cannabis use. Medical profiling via cannabinoid gene expression studies of patients who are medical candidates of cannabis could prevent the negative effects associated with its use.
... CBD has been shown to have therapeutic benefits in cancer. The analgesic effect of Cannabidiol is the result of binding to cannabinoid receptors and reduction of various types of pain: chronic (Boyaji et al. 2020;Schilling et al. 2021), acute back pain (Eskander et al. 2020), pain in patients with malignant diseases (Darkovska-Serafimovska et al. 2018). ...
Article
Full-text available
Cannabis sativa L. is a medicinal plant from family Cannabaceae with many pharmacological activities. The recent appearance of a number of cannabis products in the pharmaceutical market has led to increased requirements for regulation and quality control. The control of Cannabis sativa L. is subject to the Psychotropic Substances Convention, which includes the addictive psychoactive delta-9-tetrahydrocannabinol (THC), and to the Narcotic Drugs Convention, which includes the illicit products: herbal and liquid cannabis, resin, extracts and tinctures of flowering or fruiting tops containing THC. Approved by the FDA for use in chemotherapy are Marinol caps. (THC) and Cesamet caps., containing the synthetic THC-derivative Nabilon. There is no harmonized European Union legislation on the use of cannabis and Cannabidiol (CBD), which antagonizes the THC-psychotropic effect. Legitimate products include seeds, oil, extracts, and seed tinctures of the industrial cannabis chemotype, containing primarily Cannabidiol and less than 0.2% THC. Sativex oral spray (THC/CBD = 1:1) is approved for muscle spasticity in multiple sclerosis. Cannabidiol was not used as a food ingredient in the European Union before 15.05.1997 and is a „novel food” according to Regulation 2015/2283. Flour; protein powder and cannabis seed oil are not „novel foods”. Cannabidiol as Epidiolex is FDA approved for epilepsy forms Lennox-Gastaut and Dravet.
... A randomized controlled clinical trial (RCT) by the Portenoy group found that Nabiximols (Sativex), an oral spray consisting of Δ 9 -tetrahydrocannabinol (THC) and cannabidiol (CBD) in a 1:1 ratio, provided a small but statistically significant benefit to cancer pain (Portenoy et al., 2012). This effect was supported by reviews and meta-analyses, which also found a similar effect for cannabis and synthetic forms of THC in managing cancer pain (Darkovska-Serafimovska et al., 2018;Wang et al., 2021). However, a second set of RCTs by the Fallon group failed to find any significant difference from placebo in the management of cancer pain by Nabiximols. ...
... All studies utilized cannabis-based medication to gauge the efficacy on the symptoms under treatment and/or the safety of medicinal cannabinoids, including synthetic cannabinoids (Nabilone and Levonantradol), Nabiximol, a plant-based THC:CBD spray (Sativex) and the man-made Dronabinol. Regarding administration, 35% of the studies focused on one route, such as capsules (Abo Youssef et al., 2017;Tateo, 2017;Velayudhan et al., 2021;Wang et al., 2019;Wong et al., 2020), smoking/inhalation (Deshpande et al., 2015;Mücke et al., 2018), and oral-mucosal (Allan et al., 2018;Boland et al., 2020;Darkovska-Serafimovska et al., 2018). The rest included a range of administration routes, such as capsules and intramuscular injections (Gazendam et al., 2020;Stevens & Higgins, 2017), capsules and oral-mucosal (Aviram & Samuelly-Leichtag, 2017;Dos Santos et al., 2020;Fitzcharles et al., 2016), and capsules and smoking/inhalation (Ghabrash et al., 2020;Kafil et al., 2018;Kurlyandchik et al., 2020;Schussel et al., 2018;Wilkinson et al., 2016). ...
Article
Full-text available
Purpose: Although medicinal cannabis is prescribed for conditions such as pain, epilepsy, nausea and vomiting during cancer treatment, evidence about associated adverse side effects is still evolving. Because adverse events (AEs) might impact the performance of workers, it is important to consider their implications on workplace health and safety (WHS). This study aimed to map the types and prevalence of the AEs associated with medical cannabis and articulate how those events could impact WHS. Methods: A scoping review of systematic reviews and/or meta-analyses published between 2015 and March 2021 was performed to identify the AEs of medicinal cannabis in adults. Publications in English and full text available online were collected from Embase, MEDLINE, PsychINFO, PubMed, Scopus, and Web of Science. Results: Of 1,326 papers identified from the initial search, 31 met the inclusion criteria and were analyzed. The studies reported various AEs with the most predominant being sedation, nausea/vomiting, dizziness, and euphoria. Acute and chronic pain was the most prevalent disorder under review. Conclusions: Adverse events associated with the use of medicinal cannabis could increase workplace risks, including decreased alertness and reaction times, increased absenteeism, reduced ability to safely drive or operate machinery and an increased probability of falling. Focused research into the risk to workers and workplaces from the use of medical cannabis and related human performance impairment is urgently warranted.
... Chapman and colleagues found that though small studies using THC reported analgesic benefits with cannabinoids when added to standard opioids, more extensive trials of cannabinoids were largely negative [41]. Darkovska-Serafimovska and colleagues included only three studies: the studies reported by Dr. Johnson, the Johnson follow-up paper, and the trial reported by Dr. Portnoy's study [42,43]. The review reported a favorable conclusion based upon the secondary outcome from the Johnson study, which is quoted in the abstract. ...
Article
Full-text available
Background: Approximately 18% of patients with cancer use cannabis at one time as palliation or treatment for their cancer. We performed a systematic review of randomized cannabis cancer trials to establish a guideline for its use in pain and to summarize the risk of harm and adverse events when used for any indication in cancer patients. Methods: A systematic review of randomized trials with or without meta-analysis was carried out from MEDLINE, CCTR, Embase, and PsychINFO. The search involved randomized trials of cannabis in cancer patients. The search ended on November 12, 2021. The Jadad grading system was used for grading quality. Inclusion criteria for articles were randomized trials or systematic reviews of randomized trials of cannabinoids versus either placebo or active comparator explicitly in adult patients with cancer. Results: Thirty-four systematic reviews and randomized trials met the eligibility criteria for cancer pain. Seven were randomized trials involving patients with cancer pain. Two trials had positive primary endpoints, which could not be reproduced in similarly designed trials. High-quality systematic reviews with meta-analyses found little evidence that cannabinoids are an effective adjuvant or analgesic to cancer pain. Seven systematic reviews and randomized trials related to harms and adverse events were included. There was inconsistent evidence about the types and levels of harm patients may experience when using cannabinoids. Conclusion: The MASCC panel recommends against the use of cannabinoids as an adjuvant analgesic for cancer pain and suggests that the potential risk of harm and adverse events be carefully considered for all cancer patients, particularly with treatment with a checkpoint inhibitor.
... Neste contexto, estima-se no mundo que ocorreram 18 milhões de pessoas diagnosticadas com algum tipo de câncer e o sintoma dor associado à essa doença causa séria preocupação (Bray et al, 2018), posto que segundo a Organização Mundal da Saúde (OMS, 2017), esse sintoma é relatado em cerca de 50% de pessoas com câncer e em até 90% em sua fase avançada. Na presensça da dor, o efeito canabinóide segundo pesquisa de Darkovska-Serafimovska et al. (2018) mostrou que pacientes adultos com doenças malignas em estágio terminal mesmo utilizando doses altas de opiáceos fortes possuíam intensidade de dor maior ou igual a 4 em uma escala de 0-10. Porém, a terapia analgesica com Nabiximols (spray Sativex®), medicamento oral a base de cannabis, o qual faz a combinação de um agonista parcial de CB1 (THC) com um modulador do sistema canabidiol (CBD), teve como resultado ao final de 2 semanas relatos de redução igual ou maior que 30% na intensidade da dor em uma escala de 0-10. ...
Article
Full-text available
Este estudo visou mapear o escopo das evidências científicas sobre os efeitos terapêuticos dos canabinóides no câncer de mama. Estudo metodológico de revisão de escopo da literatura científica nas bases: Scopus, PubMed e MEDline, sendo acessadas por meio do Portal de Periódico CAPES/MEC e nas bases: Science Direct, Cochrane e Biblioteca Virtual de Saúde. Após teste e reteste uttilizando o operador conectivo booleano AND, foram aplicados na nas bases de dados os seguintes descritores: (therapeutic uses cannabis) AND (therapy) AND (breast cancer or breast neoplasms). Os resultados encontrados apontaram six estudos, apenas um do tipo quantitativo e cinco de revisão de literatura que apresentaram uso terapêutico integrativo dos canabinóides e seus benefícios para o alivio dos sintomas. Concluiu-se que há escassez de evidências científicas sobre o alivio dos sintomas e das formas de uso da terapêutica integrativa das canabinóides no câncer de mama.
... 35 Pharmacotherapy is needed to reduce it. 36 Pain management affected health outcome and patients' quality of life. 37 Providing health education can increase patient knowledge in terms of pain management. ...
Article
Full-text available
Background Breast cancer patients with post-chemotherapy had self-care deficit problems. The patient is unable to perform activities of daily living which can decrease the quality of life. One of the factors that affected self-care deficit among breast cancer patients was inadequate information. This study aimed to determine the effects of peer support psychoeducation based on experiential learning on the self-care demand. Design and methods This study used a quasi-experimental design with pretest and posttest. The independent variable of the study was the application of peer support psychoeducation based on experiential learning and the dependent variable was the self-care demand. The inclusion criteria in this study were breast cancer patients with post-chemotherapy. The exclusion criteria in this study were patients who were unconscious. We selected the experimental and control group randomly. The total sample in this study was 60 people, 30 respondents for the experiment group and 30 respondents for the control group. Peer support psychoeducation based on experiential learning was developed from Wauchope’s psychoeducational theory, Orem’s self-care, and David Kolb’s experiential learning. We used a self-report questionnaire to measure self-care demand. Data were analyzed using Wilcoxon signed rank and Mann–Whitney U tests. Results Peer support psychoeducation based on experiential learning has a significant effect on self-care demand, namely ADL ( p = 0.002), pain management ( p = 0.002), nutritional management ( p = 0.000), and rest and sleep ( p = 0.000). Conclusion Psychoeducation based on experiential learning was recommended for nursing care or physician to increase self-care demand.
Article
Full-text available
As propriedades terapêuticas da planta Cannabis sativa L. é de conhecimento popular desde os povos antigos e, devido a mudanças socioculturais, sua utilização para fins terapêuticos foi reduzida. Entretanto, nos últimos anos, a busca por pesquisas científicas sobre esses efeitos terapêuticos e o incentivo de seu uso tem crescido largamente em todo mundo. O objetivo desta revisão bibliográfica é apresentar a história da planta Cannabis com uso medicinal e evidenciar seus benefícios para a odontologia. Com base em artigos encontrados nas plataformas SciELO, Pubmed e Google acadêmico, foram reunidas informações a respeito da trajetória do uso da planta na medicina e como ela interage com o nosso organismo. Deste modo, as evidências demonstram muitos benefícios para odontologia, como tratamentos de mucosites orais, devido ao efeito antioxidante e anti-inflamatório dos derivados da planta, de doenças periodontais, também pelo potencial de combate a inflamação e pela capacidade antimicrobiana e dor orofacial, uma vez que são considerados analgésicos potentes. Em geral, esta coletânea sugere que as substâncias advindas da planta, como o canabidiol, possuem propriedades biológicas promissoras e o crescente interesse nas pesquisas sobre plantas medicinais e seus fitocompostos fomentam a divulgação e os estudos sobre as aplicações da Cannabis sativa L. em odontologia.
Chapter
The purpose of this chapter is to discuss the alternative treatment modalities for the active female with musculoskeletal pain. Alternative treatment modalities are defined as any treatment that is not considered standard clinical management, i.e., surgical or pharmacological, but other nontraditional methods that have gained popularity despite scientific controversy. Topics include heat/cold treatment, exercise therapy, neuroscience education, supplements (CBD, antioxidants, glucosamine, etc.), chiropractic spinal manipulation, acupuncture, dynamic compression, Kinesio taping, transcutaneous electrical nerve stimulation (TENS), cupping, and homeopathy. Data have been collected from various journals and different studies published on PubMed and Cochrane Library. Treatments were grouped into tiers based on currently available evidence. Available literature review best supports the utilization of thermal modality, exercise regimen, neuroscience training, and specific supplements as management options for musculoskeletal pain in active females. Other treatment modalities either lack sound supporting evidence or have only low-quality evidence to support claims of beneficial outcomes.
Article
Full-text available
Introduction: Cannabis has been used for medical purposes across the world for centuries. As states and countries implement medical and recreational cannabis policies, increasing numbers of people are using cannabis pharmacotherapy for pain. There is a theoretical rationale for cannabis' efficacy for pain management, although the subjective pain relief from cannabis may not match objective measurements of analgesia. As more patients turn to cannabis for pain relief, there is a need for additional scientific evidence to evaluate this increase. Materials and Methods: Research for this review was performed in the PubMed/National Library of Medicine database. Discussion: Preclinical studies demonstrate a narrow therapeutic window for cannabis as pharmacotherapy for pain; the body of clinical evidence for this indication is not as extensive. A recent meta-analysis of clinical trials of cannabis and cannabinoids for pain found modest evidence supporting the use of cannabinoid pharmacotherapy for pain. Recent epidemiological studies have provided initial evidence for a possible reduction in opioid pharmacotherapy for pain as a result of increased implementation of medical cannabis regimens. Conclusion: With increased use of medical cannabis as pharmacotherapy for pain comes a need for comprehensive risk-benefit discussions that take into account cannabis' significant possible side effects. As cannabis use increases in the context of medical and recreational cannabis policies, additional research to support or refute the current evidence base is essential to attempt to answer the questions that so many healthcare professionals and patients are asking.
Article
Full-text available
This article reviews recent research on cannabinoid analgesia via the endocannabinoid system and non-receptor mechanisms, as well as randomized clinical trials employing canna- binoids in pain treatment. Tetrahydrocannabinol (THC, Marinol ® ) and nabilone (Cesamet ® ) are currently approved in the United States and other countries, but not for pain indications. Other synthetic cannabinoids, such as ajulemic acid, are in development. Crude herbal cannabis remains illegal in most jurisdictions but is also under investigation. Sativex ® , a cannabis derived oromucosal spray containing equal proportions of THC (partial CB 1 receptor agonist ) and can- nabidiol (CBD, a non-euphoriant, anti-infl ammatory analgesic with CB 1 receptor antagonist and endocannabinoid modulating effects) was approved in Canada in 2005 for treatment of central neuropathic pain in multiple sclerosis, and in 2007 for intractable cancer pain. Numer- ous randomized clinical trials have demonstrated safety and effi cacy for Sativex in central and peripheral neuropathic pain, rheumatoid arthritis and cancer pain. An Investigational New Drug application to conduct advanced clinical trials for cancer pain was approved by the US FDA in January 2006. Cannabinoid analgesics have generally been well tolerated in clinical trials with acceptable adverse event profi les. Their adjunctive addition to the pharmacological armamentarium for treatment of pain shows great promise.
Article
Full-text available
Marijuana use and its distribution raise several complex health, social, and legal issues in the United States. Marijuana is prohibited in only 23 states and promarijuana laws are likely to be introduced in these states in the future. Increased access to and legalization of medical marijuana may have an impact on recreational marijuana use and perception through increased availability and decreased restrictiveness around the drug. The authors undertook an analysis to characterize the policy features of medical marijuana legislation, including an emphasis on the types of medical conditions that are included in medical marijuana laws. A high degree of variability in terms of allowable medical conditions, limits on cultivation and possession, and restrictiveness of policies was discovered. Further research is needed to determine if this variability impacts recreational use in those states.
Article
Full-text available
The Cannabinoid Use in Progressive Inflammatory brain Disease (CUPID) trial aimed to determine whether or not oral Δ(9)-tetrahydrocannabinol (Δ(9)-THC) slowed the course of progressive multiple sclerosis (MS); evaluate safety of cannabinoid administration; and, improve methods for testing treatments in progressive MS. There were three objectives in the CUPID study: (1) to evaluate whether or not Δ(9)-THC could slow the course of progressive MS; (2) to assess the long-term safety of Δ(9)-THC; and (3) to explore newer ways of conducting clinical trials in progressive MS. The CUPID trial was a randomised, double-blind, placebo-controlled, parallel-group, multicentre trial. Patients were randomised in a 2 : 1 ratio to Δ(9)-THC or placebo. Randomisation was balanced according to Expanded Disability Status Scale (EDSS) score, study site and disease type. Analyses were by intention to treat, following a pre-specified statistical analysis plan. A cranial magnetic resonance imaging (MRI) substudy, Rasch measurement theory (RMT) analyses and an economic evaluation were undertaken. Twenty-seven UK sites. Adults aged 18-65 years with primary or secondary progressive MS, 1-year evidence of disease progression and baseline EDSS 4.0-6.5. Oral Δ(9)-THC (maximum 28 mg/day) or matching placebo. Three and 6 months, and then 6-monthly up to 36 or 42 months. Primary outcomes were time to EDSS progression, and change in Multiple Sclerosis Impact Scale-29 version 2 (MSIS-29v2) 20-point physical subscale (MSIS-29phys) score. Various secondary patient- and clinician-reported outcomes and MRI outcomes were assessed. RMT analyses examined performance of MS-specific rating scales as measurement instruments and tested for a symptomatic or disease-modifying treatment effect. Economic evaluation estimated mean incremental costs and quality-adjusted life-years (QALYs). Effectiveness - recruitment targets were achieved. Of the 498 randomised patients (332 to active and 166 to placebo), 493 (329 active and 164 placebo) were analysed. Primary outcomes: no significant treatment effect; hazard ratio EDSS score progression (active : placebo) 0.92 [95% confidence interval (CI) 0.68 to 1.23]; and estimated between-group difference in MSIS-29phys score (active-placebo) -0.9 points (95% CI -2.0 to 0.2 points). Secondary clinical and MRI outcomes: no significant treatment effects. Safety - at least one serious adverse event: 35% and 28% of active and placebo patients, respectively. RMT analyses - scale evaluation: MSIS-29 version 2, MS Walking Scale-12 version 2 and MS Spasticity Scale-88 were robust measurement instruments. There was no clear symptomatic or disease-modifying treatment effect. Economic evaluation - estimated mean incremental cost to NHS over usual care, over 3 years £27,443.20 per patient. No between-group difference in QALYs. The CUPID trial failed to demonstrate a significant treatment effect in primary or secondary outcomes. There were no major safety concerns, but unwanted side effects seemed to affect compliance. Participants were more disabled than in previous studies and deteriorated less than expected, possibly reducing our ability to detect treatment effects. RMT analyses supported performance of MS-specific rating scales as measures, enabled group- and individual person-level examination of treatment effects, but did not influence study inferences. The intervention had significant additional costs with no improvement in health outcomes; therefore, it was dominated by usual care and not cost-effective. Future work should focus on determining further factors to predict clinical deterioration, to inform the development of new studies, and modifying treatments in order to minimise side effects and improve study compliance. The absence of disease-modifying treatments in progressive MS warrants further studies of the cannabinoid pathway in potential neuroprotection. Current Controlled Trials ISRCTN62942668. The National Institute for Health Research Health Technology Assessment programme, the Medical Research Council Efficacy and Mechanism Evaluation programme, Multiple Sclerosis Society and Multiple Sclerosis Trust. The report will be published in full in Health Technology Assessment; Vol. 19, No. 12. See the NIHR Journals Library website for further project information.
Article
Full-text available
Cannabinoids have been attracting a great deal of interest as potential anticancer agents. Originally derived from the plant Cannabis sativa, there are now a number of endo-, phyto- and synthetic cannabinoids available. This review summarizes the key literature to date around the actions, antitumor activity, and mechanisms of action for this broad range of compounds. Cannabinoids are largely defined by an ability to activate the cannabinoid receptors – CB1 or CB2. The action of the cannabinoids is very dependent on the exact ligand tested, the dose, and the duration of exposure. Some cannabinoids, synthetic or plant-derived, show potential as therapeutic agents, and evidence across a range of cancers and evidence in vitro and in vivo is starting to be accumulated. Studies have now been conducted in a wide range of cell lines, including glioma, breast, prostate, endothelial, liver, and lung. This work is complemented by an increasing body of evidence from in vivo models. However, many of these results remain contradictory, an issue that is not currently able to be resolved through current knowledge of mechanisms of action. While there is a developing understanding of potential mechanisms of action, with the extracellular signal-regulated kinase pathway emerging as a critical signaling juncture in combination with an important role for ceramide and lipid signaling, the relative importance of each pathway is yet to be determined. The interplay between the intracellular pathways of autophagy versus apoptosis is a recent development that is discussed. Overall, there is still a great deal of conflicting evidence around the future utility of the cannabinoids, natural or synthetic, as therapeutic agents.
Article
Laboratory evidence has shown that cannabinoids might have a neuroprotective action. We investigated whether oral dronabinol (Δ(9)-tetrahydrocannabinol) might slow the course of progressive multiple sclerosis.
Article
As a therapeutic agent, most people are familiar with the palliative effects of the primary psychoactive constituent of Cannabis sativa (CS), Δ(9)-tetrahydrocannabinol (THC), a molecule active at both the cannabinoid 1 (CB1) and cannabinoid 2 (CB2) receptor subtypes. Through the activation primarily of CB1 receptors in the central nervous system, THC can reduce nausea, emesis and pain in cancer patients undergoing chemotherapy. During the last decade, however, several studies have now shown that CB1 and CB2 receptor agonists can act as direct antitumor agents in a variety of aggressive cancers. In addition to THC, there are many other cannabinoids found in CS, and a majority produces little to no psychoactivity due to the inability to activate cannabinoid receptors. For example, the second most abundant cannabinoid in CS is the non-psychoactive cannabidiol (CBD). Using animal models, CBD has been shown to inhibit the progression of many types of cancer including glioblastoma (GBM), breast, lung, prostate and colon cancer. This review will center on mechanisms by which CBD, and other plant-derived cannabinoids inefficient at activating cannabinoid receptors, inhibit tumor cell viability, invasion, metastasis, angiogenesis, and the stem-like potential of cancer cells. We will also discuss the ability of non-psychoactive cannabinoids to induce autophagy and apoptotic-mediated cancer cell death, and enhance the activity of first-line agents commonly used in cancer treatment.
Article
Cannabis has been used in medicine for thousands of years prior to achieving its current illicit substance status. Cannabinoids, the active components of Cannabis sativa, mimic the effects of the endogenous cannabinoids (endocannabinoids), activating specific cannabinoid receptors, particularly CB1 found predominantly in the central nervous system and CB2 found predominantly in cells involved with immune function. Delta-9-tetrahydrocannabinol, the main bioactive cannabinoid in the plant, has been available as a prescription medication approved for treatment of cancer chemotherapy-induced nausea and vomiting and anorexia associated with the AIDS wasting syndrome. Cannabinoids may be of benefit in the treatment of cancer-related pain, possibly synergistic with opioid analgesics. Cannabinoids have been shown to be of benefit in the treatment of HIV-related peripheral neuropathy, suggesting that they may be worthy of study in patients with other neuropathic symptoms. Cannabinoids have a favorable drug safety profile, but their medical use is predominantly limited by their psychoactive effects and their limited bioavailability. This article is protected by copyright. All rights reserved. © 2015 American Society for Clinical Pharmacology and Therapeutics.
Article
Laboratory evidence has shown that cannabinoids might have a neuroprotective action. We investigated whether oral dronabinol (Δ(9)-tetrahydrocannabinol) might slow the course of progressive multiple sclerosis. In this multicentre, parallel, randomised, double-blind, placebo-controlled study, we recruited patients aged 18-65 years with primary or secondary progressive multiple sclerosis from 27 UK neurology or rehabilitation departments. Patients were randomly assigned (2:1) to receive dronabinol or placebo for 36 months; randomisation was by stochastic minimisation, using a computer-generated randomisation sequence, balanced according to expanded disability status scale (EDSS) score, centre, and disease type. Maximum dose was 28 mg per day, titrated against bodyweight and adverse effects. Primary outcomes were EDSS score progression (masked assessor, time to progression of ≥1 point from a baseline score of 4·0-5·0 or ≥0·5 points from a baseline score of ≥5·5, confirmed after 6 months) and change from baseline in the physical impact subscale of the 29-item multiple sclerosis impact scale (MSIS-29-PHYS). All patients who received at least one dose of study drug were included in the intention-to-treat analyses. This trial is registered as an International Standard Randomised Controlled Trial (ISRCTN 62942668). Of the 498 patients randomly assigned to a treatment group, 329 received at least one dose of dronabinol and 164 received at least one dose of placebo (five did not receive the allocated intervention). 145 patients in the dronabinol group had EDSS score progression (0·24 first progression events per patient-year; crude rate) compared with 73 in the placebo group (0·23 first progression events per patient-year; crude rate); HR for prespecified primary analysis was 0·92 (95% CI 0·68-1·23; p=0·57). Mean yearly change in MSIS-29-PHYS score was 0·62 points (SD 3·29) in the dronabinol group versus 1·03 points (3·74) in the placebo group. Primary analysis with a multilevel model gave an estimated between-group difference (dronabinol-placebo) of -0·9 points (95% CI -2·0 to 0·2). We noted no serious safety concerns (114 [35%] patients in the dronabinol group had at least one serious adverse event, compared with 46 [28%] in the placebo group). Our results show that dronabinol has no overall effect on the progression of multiple sclerosis in the progressive phase. The findings have implications for the design of future studies of progressive multiple sclerosis, because lower than expected progression rates might have affected our ability to detect clinical change. UK Medical Research Council, National Institute for Health Research Efficacy and Mechanism Evaluation programme, Multiple Sclerosis Society, and Multiple Sclerosis Trust.