Opioid-Sparing Effect of Cannabinoids For Analgesia

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

www.nature.

com/npp

ARTICLE OPEN

Opioid-sparing effect of cannabinoids for analgesia: an updated


systematic review and meta-analysis of preclinical and clinical
studies

Suzanne Nielsen1,2 , Louisa Picco1, Bridin Murnion3, Bryony Winters 4, Justin Matheson 5, Myfanwy Graham6,7,
Gabrielle Campbell8, Laila Parvaresh9,10, Kok-Eng Khor11,12, Brigid Betz-Stablein13, Michael Farrell2, Nicholas Lintzeris3,9 and
Bernard Le Foll 5,14

© The Author(s) 2022

Cannabinoid co-administration may enable reduced opioid doses for analgesia. This updated systematic review on the opioid-
sparing effects of cannabinoids considered preclinical and clinical studies where the outcome was analgesia or opioid dose
requirements. We searched Scopus, Cochrane Central Registry of Controlled Trials, Medline, and Embase (2016 onwards). Ninety-
two studies met the search criteria including 15 ongoing trials. Meta-analysis of seven preclinical studies found the median effective
dose (ED50) of morphine administered with delta-9-tetrahydrocannabinol was 3.5 times lower (95% CI 2.04, 6.03) than the ED50 of
1234567890();,:

morphine alone. Six preclinical studies found no evidence of increased opioid abuse liability with cannabinoid administration. Of
five healthy-volunteer experimental pain studies, two found increased pain, two found decreased pain and one found reduced pain
bothersomeness with cannabinoid administration; three demonstrated that cannabinoid co-administration may increase opioid
abuse liability. Three randomized controlled trials (RCTs) found no evidence of opioid-sparing effects of cannabinoids in acute pain.
Meta-analysis of four RCTs in patients with cancer pain found no effect of cannabinoid administration on opioid dose (mean
difference −3.8 mg, 95% CI −10.97, 3.37) or percentage change in pain scores (mean difference 1.84, 95% CI −2.05, 5.72); five
studies found more adverse events with cannabinoids compared with placebo (risk ratio 1.13, 95% CI 1.03, 1.24). Of five controlled
chronic non-cancer pain trials; one low-quality study with no control arm, and one single-dose study reported reduced pain scores
with cannabinoids. Three RCTs found no treatment effect of dronabinol. Meta-analyses of observational studies found 39% reported
opioid cessation (95% CI 0.15, 0.64, I2 95.5%, eight studies), and 85% reported reduction (95% CI 0.64, 0.99, I2 92.8%, seven studies).
In summary, preclinical and observational studies demonstrate the potential opioid-sparing effects of cannabinoids in the context
of analgesia, in contrast to higher-quality RCTs that did not provide evidence of opioid-sparing effects.

Neuropsychopharmacology (2022) 47:1315–1330; https://doi.org/10.1038/s41386-022-01322-4

INTRODUCTION There is substantial interest in the opioid-sparing potential of


Opioids are widely prescribed for chronic pain, but due to cannabinoids in the context of pain management. Preclinical data
concerns related to harms, recommendations have been made to have consistently demonstrated opioid-sparing effects [3]. Interest
reduce reliance on higher doses [1]. One strategy to reduce opioid from policy makers has been further driven by ecological and
dose requirements has been through use of opioid-sparing epidemiological research [4]; however, highly publicized findings
medicines. Opioid-sparing medicines can (1) delay or prevent have recently been questioned [5].
the initiation of treatment with opioid analgesics (2) decrease the The overlapping neuroanatomical distribution of opioid and
duration of opioid treatment (3) reduce the total dosages of cannabinoid receptors in the central and peripheral nervous
opioid used or (4) reduce opioid-related adverse outcomes, system in areas involved with anti-nociception support potential
without causing an unacceptable increase in pain [2]. opioid-sparing effects. Opioids and cannabinoids have comparable

1
Monash Addiction Research Centre, Eastern Health Clinical School, Monash University, Frankston, VIC, Australia. 2National Drug and Alcohol Research Centre, UNSW, Sydney,
NSW, Australia. 3Discipline of Addiction Medicine, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia. 4Pain Management Research Institute, University
of Sydney and Royal North Shore Hospital, Sydney, NSW, Australia. 5Translational Addiction Research Laboratory, Campbell Family Mental Health Research Institute, Centre for
Addiction and Mental Health (CAMH), Toronto, ON, Canada. 6The Australian Centre for Cannabinoid Clinical and Research Excellence (ACRE), Newcastle, NSW, Australia. 7Centre
Centre for Drug Repurposing and Medicines Research, School of Medicine and Public Health, The University of Newcastle, Newcastle, NSW, Australia. 8School of Health and
Behavioural Sciences, University of the Sunshine Coast, Maroochydore, QLD, Australia. 9Drug and Alcohol Services, South Eastern Sydney Local Health District, Surry Hills, NSW,
Australia. 10School of Public Health, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia. 11Medicine and Health, UNSW, Kensington, NSW, Australia.
12
Department of Pain Management, Prince of Wales Hospital, Randwick, NSW, Australia. 13The University of Queensland Diamantina Institute, The University of Queensland,
Dermatology Research Centre, Brisbane, QLD, Australia. 14Departments of Pharmacology and Toxicology, Psychiatry, Family and Community Medicine, University of Toronto,
Toronto, ON, Canada. ✉email: suzanne.nielsen@monash.edu

Received: 14 December 2021 Revised: 10 February 2022 Accepted: 31 March 2022


Published online: 22 April 2022
S. Nielsen et al.
1316
neurobiological properties with significant degree of functional checked by a second author (SN, LP, BM, JM, MG or K-EK). These same
interaction [6]. Opioid and cannabinoid receptors are Gi/o-protein- authors reviewed and resolved any inconsistencies. For abstracts without a
coupled receptors with similar intracellular signaling mechanisms, full text, and missing data, attempts were made to contact authors for
including: inhibition of the adenylate cyclase activity, reduced additional information.
activity of voltage-dependent calcium channels, activation of
inwardly-rectifying potassium channels, and stimulation of the Outcome measures
MAP kinase cascade. Cannabinoid type-1 (CB1) and mu receptors For preclinical studies, the primary outcome was the dose of opioid
can interact directly as functional heterodimers when co-expressed required to give an equivalent antinociceptive effect in the presence and
in the same neuron [7] and cannabinoid administration may absence of cannabinoids.
stimulate the synthesis and release of endogenous opioid peptides
centrally and peripherally [8]. Each of these properties would Analysis
predict a synergistic interaction between opioids and cannabi- Preclinical studies. Data were extracted and, where studies that were
noids, yet further complexity is afforded by the pharmacological sufficiently similar in design and outcome measures, meta-analysis was
profile of the drug. For example, in the case of protean agonists the undertaken. For the residual studies, a narrative review was conducted.
level of activation of cannabinoid receptors (both constitutive and To prepare the data for the meta-analysis, the ED50 and either
confidence limits or standard error were extracted from the relevant
stimulated) impacts upon the observed pharmacological effect literature. ED50 is calculated on the log10 scale. Therefore, to meet the
[9, 10], whilst partial agonists such as the endocannabinoid c 50 must be used in the meta-analysis.
assumption of normality, the log10 ED
anandamide could act as an antagonist in the presence of a more The log10 of the confidence limits must also be determined to calculate the
efficacious agonist [11]. c 50 :
standard deviation (SD) of the log10 ED
Our previous systematic review and meta-analysis found robust   c
preclinical evidence supporting the opioid-sparing potential of SD log10 EDc 50 ¼ log10 UL  log10 ED50
delta-9-tetrahydrocannabinol (THC), but limited clinical research 1:96
testing the opioid-sparing effects of cannabinoids [3]. With the where UL is the upper confidence limit.
proliferation of research in the past five years, this review aims to When only standard error was reported, the confidence limits were
provide an updated synthesis of preclinical and clinical studies on calculated using the method of Litchfield and Wilcoxon [14] and the above
the opioid-sparing effects of cannabinoids. procedure was repeated to calculate the standard deviation. This method
also allowed for the inclusion of studies that did not report exact sample
sizes for all treatment groups, as sample size was not required for the
MATERIALS AND METHODS calculation of standard deviation.
Data for the meta-analysis were analyzed using Review Manager 5.4
Search (Cochrane Collaboration, Oxford, UK). When calculating the continuous
We conducted an updated systematic literature search in accordance with
outcome of an equally effective opioid dose (e.g., the log10ED50 for
the Preferred Reporting Items for Systematic Reviews and Meta-Analyses
morphine when administered alone versus when administered with a
(PRISMA) recommendations [12]. The initial searches conducted on
cannabinoid), the inverse variance statistical method and random effects
October 29, 2015, had no date limits and the findings have been reported
model were used to compensate for study heterogeneity.
earlier, along with the methods (in lieu of a published/registered protocol)
No statistical difference was found in outcomes between the studies that
[3]. The updated searches were conducted on December 20, 2020 within
used different rodent species or nociceptive assays. Therefore, the mean
Scopus, Cochrane Central Registry of Controlled Trials, Medline, and
difference of log10 ED50 and the corresponding 95% confidence intervals
Embase databases and results were combined with the earlier search. A
were calculated. Due to the nature of log calculations, the mean difference—
combination of search terms relating to opioids (e.g., analgesics, opioid*,
when back-transformed to the original units—represents the response ratio.
opiate), cannabinoids (e.g., cannabis, sativex, nabiximol, cannabidiol,
For easier interpretation, we present the reciprocal of the response rate.
tetrahydrocannabinol) and outcomes of interest (e.g., pain, opioid sparing,
opioid dose, antinociceptive) were used, consistent with the initial search
(Appendix 1). Additional targeted searches of reference lists from identified Clinical studies. The outcomes of interest in clinical studies were: (1)
studies and review articles were conducted to find additional studies not reduction in total opioid doses, (2) reductions in pain through the addition
identified by the main searches. of a cannabinoid, (3) adverse events, and (4) evidence of abuse liability. A
broad range of study designs were considered. Where studies used
sufficiently similar methods and outcome measures, meta-analyses were
Study eligibility conducted.
Eligible studies included: (i) human or animal studies; (ii) for human studies,
controlled clinical and preclinical studies where cannabinoids were
Clinical trials. Meta-analysis for clinical trials was conducted with Revman
administered within a medical or clinical therapeutic framework and the
5.4, where medians and interquartile ranges were required to be converted
study outlined details of cannabinoid administration; (iii) documented
into means and standard deviations to allow inclusion in meta-analyses,
concurrent administration of opioids and cannabinoids; (iv) an outcome of
we used methods established by Luo et al. [15] and Wan et al. [16].
either pain/analgesia (including acute, chronic, cancer and non-cancer and
experimental pain studies) or opioid requirements/opioid-sparing.
Studies were excluded based on the following criteria: (i) wrong Observational studies
intervention (e.g., cannabinoid use not defined, no cannabinoid adminis- For observational studies, meta-analyses on proportions reporting changes
tered, non-concurrent opioid and cannabinoid use, non-therapeutic opioid in opioid dose outcomes were conducted using a random effect model in
use); (ii) wrong study design (e.g., case reports, epidemiological studies, Stata (metaprop, code available on request). A pooled prevalence was
reviews, letters without empirical data, commentary or news article); (iii) no calculated with 95% confidence intervals for each of the identified
outcome measure of interest (i.e., pain/analgesia or opioid dose); (iv) full outcomes that were comparable; (i) reduced opioid use, (ii) ceased opioid
text unavailable; (v) duplicate manuscript; (vi) abstract where full paper use. For remaining outcomes, a narrative synthesis was conducted.
published; (vii) unable to confirm eligibility details, or access required data Clinical studies were scored for quality using the Grading of
from authors (Appendix 2). Recommendations Assessment, Development and Evaluation (GRADE)
Titles and abstracts, and full texts were screened independently by two criteria [17]. Quality ratings were not applied to preclinical studies. As all
authors (SN, LMP, JM, BM, GC, MG, LP and K-EK) using Covidence software meta-analyses had less than ten studies funnel plots were not used to
[13]. Where inconsistencies were identified, the authors were able to reach assess bias [18].
consensus on each occasion.

Data extraction and outcomes RESULTS


The same data extraction forms used in the initial review were used. All Ninety eligible publications representing data from 92 studies
data were extracted by one of the authors (SN, LMP and BW, BM) and were identified; 29 in the initial searches and 63 in the updated

Neuropsychopharmacology (2022) 47:1315 – 1330


S. Nielsen et al.
1317
searches. Forty preclinical (21 since 2016) and 37 clinical studies Meta-analysis identified an opioid-sparing effect with morphine
(controlled trials n = 20 [12 since 2016] and observational n = 17 and delta-9-THC co-administration with one study [47] added to
[13 since 2016]) were identified for inclusion (see Appendix 3). the previous meta-analysis, Z = 4.46, p < 0.001 (mean difference in
Fifteen registered clinical trials, where data were not yet available log10ED50 = –0.54 [–0.78, –0.31]). As there was significant hetero-
were also identified. geneity in the data (I2 = 99%), a random effects model was used.
When back-transformed to the original units, the response ratio
Summary of preclinical studies was 3.5 (95% CI 2.04, 6.03) indicating that the median effective
Forty preclinical studies were identified in which the analgesic dose (ED50) of morphine was 3.5 times lower when administered
effect of opioid and cannabinoid co-administration was examined with delta-9-THC compared to when administered alone.
[19–58]. Sixteen of these studies examined delta-9-THC, while
smaller numbers of studies examined 20 other cannabinoids, Results from clinical studies
including agonists mixed CB1/CB2 agonists (CP55,940, WIN55,212- Thirty-five eligible publications representing 37 clinical studies
2, HU-210), CB1 agonists (ACEA, ACPA), CB2 agonists (beta- with 5180 participants provided data relevant to the research
caryophyllene, JWH-015, JWH-133, LY2828360), antagonists/ question (Table 2).
inverse agonists at the CB1 (AM-251) and CB2 receptor (JTE-907)
and other cannabinoids (AM1241, cannabinol, cannabidiol [CBD], Clinical trials—experimental pain. Five laboratory-based studies
CP 56,667, delta-8-THC, 11-hydroxy-delta-9-THC, dextronantradol, in healthy volunteers (n = 82) examined pain responses with co-
levonantradol and GP1a) (Table 1 and Appendix 4). Opioids administered opioids and cannabinoids using double-blind
examined included morphine, codeine, and other agonists at the within-patient study designs (Table 2a). Four studies examined
mu, delta or kappa opioid receptor including buprenorphine, oral dronabinol (2.5–20 mg) [59–62] and one examined smoked
etorphine, fentanyl, heroin, oxycodone, hydromorphone, metha- cannabis [63]. Inconsistent outcomes were observed; two studies
done, LAAM, meperidine, pentazocine, spiradoline, tramadol, and found evidence of increased pain, two found some measures of
SNC80. Most studies used rodents; however, three used rhesus decreased pain, and one study found effects of cannabinoids on
monkeys and one used guinea pigs. The most common pain “unpleasantness” but not pain ratings. One study found low
antinociceptive assays were of thermal nociception although dose dronabinol (2.5 mg) decreased the analgesic effects of
assays of mechanical and chemical nociception were also utilized. oxycodone as measured with a pressure algometer with no effect
Evidence of opioid-sparing effects or synergism were found for of 5 or 10 mg dronabinol on analgesic outcomes [61]. Another
all mixed CB1/CB2 agonists (CP55,940, delta-9-THC, HU-210, study noted potentially hyperalgesic effects of cannabinoids [59].
WIN55,212–2). Morphine-induced analgesia increased with the This was in contrast to the analgesic effect observed on pain
CB1 selective agonist ACEA, though the effect was additive as threshold and tolerance with a cold pressor test when smoked
opposed to synergistic [40]. In contrast, the CB1 selective agonist cannabis was administered with 5 mg oxycodone compared
ACPA, and DAMGO (selective mu agonist) appeared to act oxycodone or cannabis alone, although effects were not found
antagonistically when administered together in a model of on measure of outcomes of pain intensity or bothersomeness [63].
mechanical hyperalgesia [41]. The CB1 antagonist/inverse agonist Dunn et al. [62] demonstrated analgesic effects from dronabinol
AM-251 reduced the analgesic effect of morphine [40]. Conflicting 2.5 mg when co-administered with hydromorphone on thermal
outcomes were seen for CB2 selective agonists (some evidence of pain measures, but not with higher doses of dronabinol, or on
opioid-sparing effects for GP1a, JWH-015, LY2828360, but not for other measures of pain. Roberts et al. [60] found that the co-
beta-caryophyllene or JWH-133). JTE-907 (a CB2 antagonist) and administration of dronabinol and morphine resulted in reduced
cannabinoids with more complex pharmacology (CBD and pain “unpleasantness” compared to either drug alone. Three
cannabinol) did not demonstrate opioid-sparing effects. Three experimental studies included measures of abuse liability, and
less well characterized phytocannabinoids, including delta-9-THC found that smoked cannabis and dronabinol may increase the
metabolites, also showed evidence of synergy or opioid-sparing abuse liability ratings of oxycodone and hydromorphone using
effects (delta-8-THC, 11-hydroxy-delta-9-THC and levonantradol), measures such as ratings of feeling high and drug liking [61–63].
while no opioid-sparing effects were seen for other less well
characterized cannabinoids (CP, 56,667 and dextronantradol). Clinical trials—acute pain. Three double-blind randomized con-
trolled trials (n = 545) examined the opioid-sparing effects of CBD
Measures of abuse liability. Six studies reported on measures of in acute pain [64–66]. Nabilone and dronabinol were examined in
abuse liability including intracranial self-stimulation (ICSS) [38], acute post-operative pain and CBD in acute low back pain
conditioned place preference [43, 44], oxycodone self- (<30 days duration). No benefit on opioid dose requirements or
administration [50], and drug discrimination [32, 33]. None analgesic outcomes was identified (Table 2b).
provided evidence that cannabinoids increased abuse liability.
CP55,940 had no effect on ICSS with morphine or tramadol [38], Clinical trials—cancer pain. Seven controlled trials (1795 partici-
JWH105 when co-administered with morphine reduced condi- pants) investigated the opioid-sparing effect of cannabinoids in
tioned place preference, and LY2828360 when administered with patients with different forms of cancer pain. One small, non-
morphine blocked condition place preference [43, 44]. THC randomized study found a non-significant effect of cannabis on
reduced oxycodone self-administration [50], and attenuated the pain control [67], and a second pilot found no effect of medical
discriminative stimulus effect of morphine and heroin in cannabis on pain, but an increase in opioid dose in a group that
nondependent monkeys, but not in dependent monkeys [33]. received delayed cannabis [68] (Table 2c). The remaining studies
CP55,940 and WIN55,212 reduced the discriminative stimulus were all larger single or double-blind randomized trials. Five
effect of morphine and decreased heroin self-administration, both randomized controlled trials (reported in four publications)
effects were reversed by the CB1 receptor inverse agonist examined THC and nabiximols compared to placebo in patients
rimonabant [32]. with cancer pain who were taking opioids [69–72]. Two studies
found improved analgesia with nabiximols compared to the
Meta-analysis of preclinical studies. Seven studies used suffi- placebo. Johnson et al. [69] found no effect of nabiximols on
ciently similar approaches to enable a meta-analysis [19–24, 47] breakthrough opioid dose requirements. Portenoy et al. [70]
(Fig. 1). All studies included in the meta-analysis used rodents and conducted a dose-ranging study, and a significant analgesic effect
reported comparable antinociceptive doses of morphine alone was only found in the lowest dose group, with poorer tolerability
and morphine co-administered with delta-9-THC. observed for higher doses. The remaining three studies found no

Neuropsychopharmacology (2022) 47:1315 – 1330


S. Nielsen et al.
1318
Table 1. Summary of opioid-sparing outcomes in preclinical studies by cannabinoid type.
Cannabinoid type Potential synergism/opioid-sparing effects Opioid-sparing effect not clearly
observeda or tested
Mixed CB1/CB2 agonists
CP55,940 (mixed CB1/CB2 agonist) Evidence of opioid-sparing effect: Evidence of synergy/opioid-sparing
Alsalem et al. 2019 (morphine “potential synergy” not found:
mechanical nociception) Alsalem et al. 2019 (tramadol, mechanical
Maguire and France 2018 (morphine, thermal nociception)
nociception); Welch 1992 (morphine, thermal
Maguire 2013 (Rhesus monkey, morphine, thermal nociception);
nociception) Maguire and France 2016 (spiradoline,
thermal nociception)
Maguire and France 2018 (etorphine,
thermal nociception);
Minervini 2017 (spiradoline, thermal
nociception)
Delta-9-THC (partial agonist CB1/CB2) Evidence of opioid-sparing effect: Evidence of synergy/opioid-sparing
Cox 2007 (morphine, mechanical nociception) not found:
Cichewicz 2005 (guinea pigs, fentanyl and Maguire and France 2018 (etorphine,
buprenorphine, mechanical nociception) thermal nociception)
Maguire and France 2018 (morphine, thermal Opioid-sparing/synergism not directly
nociception) tested:
Nguyen 2019 (oxycodone “possibly synergistic, Wakley 2011—synergism not tested,
thermal nociception) (mechanical nociception)
Nilges 2020 (Rhesus monkeys, heroin, thermal Reche 1996—only one dose of morphine
nociception)) examined (thermal nociception)
Cichewicz 1999 (range of opioid agonists, thermal
nociception)
Cichewicz 2003 (morphine and codeine, thermal
nociception)
Li 2008 (Rhesus monkey, morphine, thermal
nociception))
Pugh 1996 (morphine, thermal nociception)
Smith 1998 (morphine, thermal nociception)
Smith 2007(morphine, thermal nociception)
Welch 1992(morphine, thermal nociception)
Williams 2006 (codeine and morphine, thermal
nociception)
Williams 2008 (morphine, thermal nociception)
HU-210 (mixed CB1/CB2 agonist) Evidence of potential opioid-sparing effect: Evidence of synergy/opioid-sparing
Sierra 2019 (SNC80 [delta opioid agonist] mechanical not found:
nociception with neuropathic pain model) Alsalem et al. 2020 morphine and
tramadol, mechanical nociception)
Wilson 2008 (morphine, thermal
nociception)
WIN55,212–2 (mixed CB1/CB2 agonist) Evidence of opioid-sparing effect: Evidence of synergy/opioid-sparing
Alsalem et al. 2020 (tramadol mechanical not found:
nociception); Alsalem et al. 2020 (not morphine,
Chen et al. 2019 (morphine, thermal nociception and mechanical nociception)
formalin)
Yesilurt 2003 (morphine, thermal nociception)
CB1 selective agonist
ACEA Evidence of synergy/opioid-sparing
not found:
Altun 2015 (morphine, thermal
nociception)
ACPA Evidence of synergy/opioid-sparing
not found:
Auh et al. 2016 (DAMGO, mechanical
nociception)
CB1 antagonist/inverse agonist
AM-251 (also has agonist activity Evidence of synergy/opioid-sparing
at GPR55) not found:
Altun 2015 (morphine, thermal
nociception)
CB2 selective agonist
JWH-015 Evidence of opioid-sparing effect: Evidence of synergy/opioid-sparing
Grenald et al. 2017 (morphine, mechanical and not found:
thermal nociception, formalin pain assay)

Neuropsychopharmacology (2022) 47:1315 – 1330


S. Nielsen et al.
1319
Table 1. continued
Cannabinoid type Potential synergism/opioid-sparing effects Opioid-sparing effect not clearly
observeda or tested
Altun 2015 (morphine, thermal
nociception)
Beta-caryophyllene Evidence of synergy/opioid-sparing
not found:
Katsuyama 2013 (morphine, capsaicin
pain assay)
JWH-133 Evidence of synergy/opioid-sparing
not found:
Yuill 2017 (morphine, noxious stimuli)
LY2828360 Evidence of opioid-sparing effect:
Iyer 2020 (morphine, mechanical nociception)
CB2 complex
AM1241 (Protean agonist CB2) Evidence of potential synergy: Evidence of synergy/opioid-sparing
Zhang 2018 (morphine, thermal and mechanical not found:
nociception) Stachtari 2016 (tramadol, thermal
nociception)
Zhang 2017 (morphine, thermal
nociception);
Zhang 2016 (morphine, thermal and
mechanical nociception)
GP1a (CB2 agonist/inverse agonist) Evidence of potential opioid-sparing effect: Evidence of synergy/opioid-sparing
Chen et al. 2019 (morphine, thermal nociception) not found:
Chen et al. 2019 (morphine, formalin
pain assay)
CB2 antagonist
JTE-907 (CB2 antagonist) Evidence of synergy/opioid-sparing
not found:
Altun 2015 (morphine, thermal
nociception)
Complex
CBD (inverse agonist/NAM CB1, partial Evidence of synergy/opioid-sparing
agonist CB2) not found:
Neelakantan 2015 (morphine, thermal and
chemical nociception)
Cannabinol (partial CB1 inverse agonist Evidence of synergy/opioid-sparing
or agonist at CB2) not found:
Nilges 2020 (heroin, thermal nociception)
Less characterized cannabinoids
CP 56,667 Evidence of synergy/opioid-sparing
not found:
Welch 1992 (morphine, thermal
nociception)
Delta-8-THC Evidence of opioid-sparing effect:
Welch 1992 (morphine, thermal nociception)
11-hydroxy-delta-9-THC Evidence of opioid-sparing effect:
Welch 1992 (morphine, thermal nociception)
Dextronantradol Evidence of synergy/opioid-sparing
not found:
Welch 1992 (morphine, thermal
nociception)
Levonantradol Evidence of opioid-sparing effect:
Welch 1992 (morphine, thermal nociception]
Species were rodents unless otherwise specified; full study details provided in Supplementary material (see Appendix 3).
a
Where opioid-sparing effect reported as not observed, results were additive rather than synergistic; or no increased analgesic effect was observed.

benefit of adding cannabinoids on their primary outcome of found [69–72]. No cancer pain studies included measures of abuse
analgesia. Although Lichtman et al. [72] did not find a significant liability.
effect of cannabinoids on pain in an intention to treat analysis, the Meta-analyses were possible on the outcomes of change in
per-protocol analysis did find a significant effect (Table 2c). Four of mean total oral morphine equivalent daily dose (OMEDD) from
seven studies required maintenance opioid doses to be kept baseline (n = 4 studies), percent change in pain score from
stable [70–72]; five studies measured breakthrough opioid doses baseline (n = 4 studies) and adverse events (n = 5 studies). Meta-
requirements as an outcome with no evidence of a difference analysis of four studies (n = 1119 participants) found no effect of

Neuropsychopharmacology (2022) 47:1315 – 1330


Table 2. Clinical studies. 1320
Study Study design Population Observation period Opioid used Cannabinoid Used Comparator Effect of cannabinoid on Outcome on analgesia GRADE evidence
reference opioid dose observed rating and other
notes
a. Experimental pain studies
Babalonis 2019 Within-subject crossover, Healthy volunteers Nine outpatient Oxycodone 0, 5, Dronabinol l 0, 2.5, 5 Placebo Cold pressor test: 2.5 mg Pressure algometer: GRADE rating
randomized, double-blind (n=10), aged 18–50 experimental sessions 10 mg (oral) mg (oral) dronabinol dronabinol + 5 mg Dronabinol a 2.5 mg dose “moderate”,
placebo-controlled years, without acute (8.5 h/session) with a capsules and oxycodone decreased decreased the analgesic placebo-controlled
design. Analgesia or chronic pain minimum of 48 h placebo tolerance (17.9 [±2.4] s), effects of 10 mg blinded study,
assessed with cold conditions and no separating each oxycodone compared with the 5 mg oxycodone (no effect indirect evidence as
pressor, pressure recent opioid or session, dronabinol tablets oxycodone dose alone from 5 mg dronabinol). use of experimental
algometer, hot thermode, cannabinoid use. Six administered 1 h (34.3 [±17.7] s) No effect on other pain pain.
cold hyperalgesia female, mean age of before oxycodone, measures (pressure Dronabinol
26.3 years pain measures up to algometer, cold pressor increased abuse
6 h after dronabinol test and hot thermode) liability ratings of
administration oxycodone
Cooper 2018 Within-subject, Healthy volunteers (n 6 outpatient Oxycodone 0, 2.5 Cannabis cigarettes Placebo Cannabis and low dose of Cannabis and 2.5 mg and GRADE rating
randomized, placebo- = 18) 21–45 years, experimental or 5.0 mg (oral) (0.0 or 5.6% THC cannabis oxycodone (2.5 mg) did 5 mg oxycodone “moderate”,
controlled, double-blind with who current sessions. Placebo or content); Participants cigarettes (0% not elicit analgesia on increased pain threshold placebo-controlled
study Analgesia was cannabis use. Six oxycodone was smoked 70% of an THC); Placebo their own; when on cold pressor test blinded study,
assessed with cold (33%) female, mean administered 45 min 800 mg cannabis oxycodone administered together, compared to the indirect evidence as
pressor test age 29.9 years before cannabis. cigarette (CBD capsules pain (with cold pressor cannabis alone (p < 0.05). use of experimental
Observations for 5 h content not stated) test) was significantly Mean reductions from pain
after cannabis reduced, pointing to the pain (McGill Pain Smoked cannabis
administration; opioid-sparing effects Questionnaire) Placebo increased
repeated pain 2.2 ± 0.5; THC alone 1.5 ± subjective abuse
assessments until 3 h. 0.5; 2.5 mg OXY 2.0 ± 0.5; liability measures
72 h washout 2.5 mg OXY + THC; 0.7 ± for oxycodone
between sessions 0.6; 5 mg OXY 1.7 ± 0.4; 5
mg OXY + THC 1.2 ± 0.4.
Pain Intensity and
bothersomeness Scales
did not differ between
cannabis, oxycodone, the
combination or placebo
Dunn 2021 Double-blind, within- Healthy adults (n = Five outpatient Hydromorphone Dronabinol 2.5, 5.0, Placebo Opioid dose held Limited evidence of GRADE rating
subject randomized, 29) with no history of laboratory sessions 4 mg (oral) 10 mg; (oral) hydromorphone constant across all dronabinol enhancement “moderate”, indirect
S. Nielsen et al.

placebo-controlled, drug use disorder, (min. 7 days apart). (no placebo sessions of hydromorphone on evidence as use of
human laboratory study 52% female, mean Sessions lasted 8 h. dronabinol pain measures. experimental pain.
using quantitative 30.4 years Study drugs co- condition) Dronabinol 2.5 mg had a Higher doses of
sensory testing measures administered at with significant effect of dronabinol (5 mg
of acute (thermal, hourly pain thermal threshold and and 10 mg) also
pressure pain; thermal, assessments for 4 h tolerance. Most pain showed greater
punctate probe temporal measures did not show a evidence of
summation; cold pressor; significant difference potential for abuse
conditioned pain between dronabinol+ and adverse effects
modulation) and chronic hydromorphone and
pain (capsaicin 10% hydromorphone alone.
topical cream with No dose effect with
thermal rekindling) dronabinol
Naef 2003 Experimental heat, cold, Healthy cannabis Four study sessions Morphine 30 mg Dronabinol 20 mg Matched No significant analgesic Potentiation of analgesia GRADE rating
pressure, single and naïve volunteers (n = with at least seven (oral) (oral) placebo capsule effect of dronabinol or not observed in this “moderate”, indirect
repeated transcutaneous 12), 6 female, mean days washout compared with morphine-dronabinol experimental pain study, evidence as use of
electrical stimulation age 25 years between sessions. THC alone, combination on heat, potential hyperalgesic experimental pain
pain, randomized, Study medications morphine alone pressure and cold tests. effect of cannabinoids
placebo-controlled, co-administered, with or THC- Additive effect of noted which may reduce
double-blind, within- pain measurements morphine morphine on analgesic effects of
subject study hourly for up to 8 h combination transcutaneous electrical morphine
stimulation test
Roberts 2006 Experimental thermal Healthy volunteers (n Four lab sessions; Morphine 0.02 Dronabinol 5 mg Placebo Not applicable (opioid Combination of GRADE rating
pain. Double-blind, four = 13) with no recent Dronabinol mg/kg (oral) dronabinol dose held constant) dronabinol and morphine “moderate”,
treatment within-subject opioid or cannabinoid administered, 90 min intravenous (1.4 capsule and did not have effect on placebo-controlled
design use. Six female, aged later morphine mg dose for 70 placebo pain intensity. The blinded study,
18–49 years administered, thermal mg adult) (i.e., morphine combination was indirect evidence as
pain measured 15 sub- analgesic) injection (normal reported to have a use of experimental
min after morphine saline) synergistic effect on pain.
administration affective response to pain Noted difficulties
(unpleasantness) with extrapolation
compared with either to clinical practice
drug alone (p = 0.012)
b. Controlled trials acute pain
Bebee 2021 Randomized, double- Adults with acute 48 h Oxycodone (5 mg CBD 400 mg (oral) Color matched 31/50 patients in the CBD Mean pain scores at 2 h GRADE rating
blind, placebo-controlled (<30 days duration) every 6 h, with placebo group and 27/50 in the were similar for the CBD “high”
clinical trial non-traumatic lower additional rescue prepared placebo group required (6.2 points; 95% CI,
(ACTRN12618000487213) back pain (n=100). dosing as (medium chain oxycodone. Total 5.5–6.9 points) and
Median age 47 years, required) triglyceride oil) oxycodone dose in the placebo groups (5.8
44% female CBD group was 230 mg points; 95% CI: 5.1,6.6
compared with 215 mg in points; absolute
the placebo group difference, –0.3 points;

Neuropsychopharmacology (2022) 47:1315 – 1330


95% CI, –1.3–0.6 points)
Table 2. continued
Study Study design Population Observation period Opioid used Cannabinoid Used Comparator Effect of cannabinoid on Outcome on analgesia GRADE evidence
reference opioid dose observed rating and other
notes
Levin 2017 Single-center randomized Patients scheduled for 300 min or until Specific opioid Nabilone 0.5 mg Matched Morphine equivalents No differences in pain GRADE rating
double-blind controlled elective surgery discharge from post- not reported, (oral) placebo capsule (mg) given in operating score (out of a possible “high”
trial (NCT02115529) under general anesthesia care unit converted into room: Nabilone 21.3 (SD 10) between groups;
anesthesia who had a OMEDD 15.2) vs placebo 20.0 (SD Maximum pain score (at
preoperative risk of 13.4), p = 0.40 rest) Nabilone 3.17 (SD
post-operative nausea Morphine equivalents 3.15) vs placebo 3.68 (SD
or vomiting (n = 340). (mg) post-surgery: 3.25), p = 0.43
Mean age 49.8 years, Nabilone 5.8 (SD9.2) vs Maximum pain score
100% female placebo 5.4 (SD 6.9) p = (with movement)
0.62 Nabilone 3.34 (SD 3.30) vs
placebo 4.0 (SD 3.33), p =
0.92
Seeling 2006 Randomized double-blind Prostate cancer From the day prior to Pitrimide Eight doses of Placebo Median dose of pitrimide The median of the resting GRADE rating
controlled trial (two patients <70 years, (all surgery to 2 days Dronabinol 5 mg dronabinol alone was 74 mg (IQR = pain was not different “high”, patients
groups) male) undergoing post-operation (oral) over 48 h capsules 44–90 mg) compared between the groups (p > administered their
surgery (n = 105). 53 (perioperatively) with 54 mg (IQR = 46–88 0.1) own opioid doses
in intervention and 52 mg) when administered
in control groups with dronabinol
c. Controlled trials cancer pain

Neuropsychopharmacology (2022) 47:1315 – 1330


Fallon 2017aa Study 1: multisite Adults (n = 399) with 49 days (2 weeks after Reported as 1:1 nabiximols Placebo oral No effect of nabiximols No differences in median GRADE rating
(patients at 101 centers in advanced incurable medication ceased) OMEDD Patients (Sativex®) oral mucosal spray on; total OMEDD −9.35, percent improvement “high”
12 different countries) cancer, unalleviated were recruited on mucosal spray (THC 95% CI-18.81–0.012 (p from baseline average Significant effect of
randomized, double- by an optimized “stable opioid 27 mg/mL: CBD 25 =0 .053); maintenance pain NRS score: nabiximols on
blind, placebo-controlled opioid therapy. Mean therapy”, not mg/mL) or matching OMEDD −3.63, 95% CI nabiximols 7.2% vs Subject Global
trial (NCT01361607) age 59.8 years, 49% more than 500 placebo. 14-day −10.80, 3.55 (p = 0.321); placebo 9.5% (median Impression of
female mg OMEDD titration (3–10 spray/ breakthrough OMEDD difference=−1.84%; Change, Patient
day to max tolerated −4.17, 95% CI −8.76, 0.42 confidence interval (CI): Satisfaction
dose), 35 days of (p = 0.075). (note patients −6.19%, 1.50%; p = 0.274) Questionnaire and
medication provided instructed to continue Median treatment effect Physician Global
in total pain medication) −0.02; 95% CI: −0.42, Impression of
0.38; p = 0.917) Change
Fallon 2017ba Study 2: 2-part enriched Adults (n = 406 49 days (2 weeks after Reported as Nabiximols (Sativex®) Placebo oral No effect of nabiximols Mean average pain scores GRADE rating
enrollment with entered and 206 medication ceased) OMEDD. Patients oral mucosal spray mucosal spray on: total OMEDD −7.1, increased from 3.2 to 3.7 “moderate”,
randomized withdrawal randomized) with were recruited on (THC 27 mg/mL: CBD 95% CI:23.9,9.7 (p = in the nabiximols group selection bias
design (“responders” advanced incurable “table opioid 25 mg/mL). 10-day 0.405); maintenance and 3.1 to 3.6 in the introduced through
randomized). Single-blind cancer, unalleviated therapy”, not titration, OMEDD −8.9, 95% placebo group, i.e., a enrichment design
titration for 10 days by an optimized more than 500 “Responders” (15% CI:19.7, 1.8 (p = 0.104); worsening of equal
followed by double-blind opioid therapy, mean mg of OMEDD pain score Breakthrough OMEDD severity in both
randomization to Sativex age 61.5 years, 43% improvement) 1.8, 95% CI:10.3, 14.0 (p = treatment groups
or placebo female randomized. 0.769). (note patients (estimated treatment
(NCT01424566) Treatment for 5 instructed to continue effect −0.02; 95% CI:
weeks pain medication) −0.42, 0.38; p = 0.917)
S. Nielsen et al.

Johnson 2010a Multicenter, randomized, Adults with cancer 2 weeks Varied opioids Placebo oral Patients allowed to use Change in pain score (out GRADE rating
double-blind, placebo- pain (n = 177), with reported as mg mucosal spray breakthrough medication of 10) in favor of THC:CBD “high”, placebo-
controlled, parallel-group inadequate analgesia OMEDD (IQR) (Baseline as needed; no change in compared with placebo controlled and
trial. (NCT00674609) despite chronic OMEDD for median amount of (−1.37 (p = 0.014)); the randomized.
opioid dosing. Mean 120 (50–213) THC 2.7 mg spray; placebo group breakthrough opioid THC group change was No significant
age 60.2 years, 56% mean of 8.47 sprays 120 (40–240)) medication in any group. not significant (−1.01 (p group differences
female day Mean change in opioid = 0.245)). The proportion were found in sleep
80 (30–180) THC 2.7 mg:CBD 2.5 dose from baseline of patients achieving a quality or nausea
mg per spray; mean Placebo −41.4 (SD 30% reduction in pain scores or the pain
sprays 9.26/day 201.27), THC: 36.8 (SD was 43% (23/48) in the control assessment
152.00); THC:CBD −3.5 THC:CBD group, 23% in
(SD 108.44). Median the THC group (12/45)
change in all groups 0 mg and 21% in the placebo
group (12/51)
Lichtman Randomized, multisite Adults (n = 397) with 50 days if not Specific opioids Nabiximols (Sativex®) Placebo oral Nabiximols did not Average pain score from GRADE rating
2018a double-blind, placebo- advanced cancer- entering extension not stated. Mean (THC 27 mg/mL: CBD mucosal spray impact maintenance baseline to end of “moderate”, unclear
controlled study (12 related chronic pain study total daily opioid 25 mg/mL), titration OMEDD (Estimated treatment (primary blinding and
countries) (NCT01262651) unalleviated by use at baseline 14 days, treatment difference endpoint): non- randomization.
optimized opioid 192.9 (SD 130.7) maintenance 21 [ETD] 1.46, 95% CI: −4.67, significant: 10.7% median Nabiximols was also
therapy. Mean age in the nabiximols days. Max 10 sprays 7.60, p = 0.64), improvement with associated with
59.9 years, 46% group and 186.1 per day breakthrough OMEDD nabiximols compared to greater
female (SD 131.0) in the (ETD −1.84, 95% CI: 4.5% with placebo (p = improvements than
placebo group −6.33, 2.66, p = 0.42) or 0.08). Nabiximols did not placebo in score on
total OMEDD (ETD −0.34, improve average pain the Subject Global
95% CI: −8.26, 7.28, p = NRS (p = 0.25) or worst Impression of
0.93). Protocol stated that pain NRS score (p = 0.68). Change, Physician
medications including Prespecified per-protocol Global Impression
opioids, should have analysis favored of Change, and
been continued at stable nabiximols over placebo Patient Satisfaction
doses if possible (p = 0.04) Questionnaire
Lissoni 2014 Two groups (not Adults (n = 26) with Not stated Oxycodone, Cannabis flos (19% Melatonin 5/12 (42%) achieved The number that GRADE rating “low”,
randomized): untreatable median dose of THC) was given as 20–100 mg control of pain without achieved pain control no-randomization,
1321
Table 2. continued 1322
Study Study design Population Observation period Opioid used Cannabinoid Used Comparator Effect of cannabinoid on Outcome on analgesia GRADE evidence
reference opioid dose observed rating and other
notes
cannabinoid tincture or metastatic solid 30 mg (10–60 infusion 100 ml (500 opioid dose increase was not significantly no allocation
melatonin (clinical trial tumor (age and mg), twice /day mg/L water) three compared to the control different between groups concealment
registration not reported) gender not stated) times a day group where 2/14 (14%) (5/12 in cannabis group described, greater
achieved pain control and 2/5 in melatonin) disease progression
in the cannabis
group
Portenoy Randomized, 4-arm Adults (n = 360) with 5 weeks of Median OMEDD Nabiximols 1–4 Placebo oral No change in median OR 1.87 (p = 0.038) GRADE rating
2012a double-blind, placebo- advanced cancer and medication 120 Sprays mucosal spray amount of breakthrough compared with placebo “high”. Opioid
controlled, graded-dose opioid-refractory administration (Median OMEDD opioid medication in any composite measure
study (NCT00530764) pain, mean age 58 Median OMEDD Nabiximols 6–10 120 mg) group. Note that patients OR 1.70 (p = 0.079) showed better
years, 48% female 120 Sprays were instructed to keep compared with placebo improvements in
Median OMEDD Nabiximols 11–16 opioid dose constant so OR 1.16 (p = 0.622) low dose group.
180 Sprays opioid-sparing effect on compared with placebo Lower tolerability of
opioid dose could not be THC:CBD in higher
observed dose groups
Zylla 2021 Pilot randomized Adults (n = 30) with 3 months Opioid type not Maintenance dose of Early versus late EC group had stable Mean pain scores GRADE rating “low”
controlled trial comparing stage IV cancer specified. OMEDD 30–40 mg of THC start cannabis opioid use; 3/9 in EC remained similar small sample with
early cannabis (EC) to requiring opioids measured using and 30–40 mg of group and 4/9 in DC between the two groups high attrition. Also
delayed start cannabis Patients in the EC daily diary CBD per day, titrating group increased OMEDD examined dosing
(DC) group were similar to up over 2–4 weeks by ≥20%. Three patients patterns: THC per
DC group with in the EC group patient each month
respect to mean age decreased their daily was nearly twice
(57 (SD = 9) years vs OMEDD by ≥20% that of CBD
55 (SD = 13) years) (average 34.3 mg
and percentage THC vs 16.6 mg
female (47% vs 53%), CBD)
respectively
d. Controlled trials: chronic non-cancer pain
Abrams 2011 Clinical laboratory study Adults (n = 24) 5 days Morphine (mean Vaporized cannabis No comparator NA (opioid dose held Pain score: reduction GRADE rating “low”.
of self-reported pain receiving chronic dose 62 mg, n = dose of 0.9 g of (single-arm constant) from 34.8 (95% CI: 29.4, No control arm,
under observed opioid treatment 13) or oxycodone 3.56% delta-9-THC or study) 40.1) on baseline to 24.1 placebo effects
conditions (also (mixed pain (mean dose 53 as much as they (95% CI: 18.8, 29.4) on day cannot be
measured conditions). Mean age mg, n = 11) could tolerate, 5 with morphine; and excluded. No
S. Nielsen et al.

pharmacokinetic effects 45.1 years, 48% administered three from 43.8 (95% CI: 38.6, pharmacokinetic
of concurrent female times a day 49.1) on baseline to 33.6 interaction
administration) (95% CI: 28.5, 38.6) on day observed. Cannabis
(NCT00308555) 5 with oxycodone. inhalation
Significant reduction produced a
overall subjective “high”
Abrams 2020 Randomized double- Adults with sickle cell 5 inpatient days with Hydromorphone, Cannabis plant Vaporized The mean (SD) difference The mean (SD) difference GRADE rating “low”;
blind, two group disease with chronic 30-day washout oxycodone, material containing placebo in log OMEDD dose in pain rating assessment small sample size
crossover design pain (n = 23), 21 of followed by another 5 hydrocodone, 4.4% THC and 4.9% cannabis between the cannabis using the VAS data and unclear
(NCT01771731) whom were using inpatient days morphine sulfate, CBD which were and placebo periods in between the active and blinding
opioids. Mean age, fentanyl, vaporized and this value was not placebo groups were not procedures and
37.6 years; 56% methadone and inhaled 3 times per significant (2.05 [0.21] vs significant on any day crossover design
female oxymorphone day 2.09 [0.22]; p = 0.20)
De Vries 2016 Randomized, single-dose, Adults aged 18 and 6h Pethidine; Dronabinol 8 mg Diazepam 10 mg The pharmacokinetic Primary analysis showed GRADE rating
double-blind, placebo- above with chronic tramadol and parameters of THC were no treatment effect of “moderate”
controlled, two-way abdominal pain from codeine (patients’ similar between opioid THC. When only patients downgraded due to
crossover study chronic pancreatitis usual analgesic and non-opioid users. on opioids were small sample size
(NCT01318369) (n = 24, 12 of whom medication) Opioid dose considered, the mean and crossover
were taking opioids). requirements were not an VAS pain score at 2 h was design.
Mean age of sample outcome of the single- similar for patients on in Additional data
52 years, 9 of 24 dose study THC arm (2.917, SD 2.205) provided by
patients were female and the diazepam (active authors
placebo) arm (2.53, SD
1.702)
De Vries 2017 Randomized, single-dose, Two clinical trials 61 days Codeine, Dronabinol tablet, Matched Not reported. Patients Primary analysis (all GRADE rating
double-blind, placebo- where the samples tramadol, increased to 8 mg dronabinol were asked to continue patients) VAS mean “moderate”, small
controlled, two-way were combined: (1) oxycontin, three times a day placebo tablet taking their medications scores did not differ sample size and
crossover study Adults with painful fentanyl, and over 10 days, with (including analgesics) between THC and high attrition in the
(NCT01562483 and chronic pancreatitis morphine the option to reduce according to prescription placebo. For patients on active arm for the
NCT01551511) (CP) n = 23, and (2) (patients usual to 5 mg if 8 mg is not opioids: (1) CP group (n CP group.
adults with chronic medicines) tolerated. Those not = 20) mean VAS on day Additional data
postsurgical tolerating 5 mg three 49 was 2.05, SD 2.65) with provided by
abdominal pain (PSP), times a day were THC compared with 2.94, authors
n = 27, mean age 52.9 withdrawn SD 2.10 for placebo (p =
years, 50% female 0.4). (2) PSP (n = 17),
mean VAS on day 49 for
placebo was higher (5.51,
SD 2.27) compared with
THC (2.9, SD 2.35), p =
0.03
Narang 2008 Phase 1: randomized, Adults taking opioids OMEDD mean Dronabinol 10 and Matched One subject took rescue In single-dose studies 10 GRADE rating
single-dose, double-blind, for chronic pain; BPI ≥ 68.1 mg (SD: 57.2, 20 mg placebo pain medication in all 3 and 20 mg dronabinol “moderate”,

Neuropsychopharmacology (2022) 47:1315 – 1330


Table 2. continued
Study Study design Population Observation period Opioid used Cannabinoid Used Comparator Effect of cannabinoid on Outcome on analgesia GRADE evidence
reference opioid dose observed rating and other
notes
placebo-controlled, 4 (n = 30). Pain range 7.5–228) dronabinol conditions (placebo, significantly increased randomized and
crossover trial. Primary diagnosis: Phase 1: Three, 8-h (mix of capsule dronabinol 10 mg, and the amount of analgesic placebo-controlled,
lab sessions with
outcome measure Total neuropathic (N = 7), three days washout. oxycodone, dronabinol 20 mg), one relief reported. Total pain small sample
Pain Relief score (Phase 2 nociceptive (N = 7), morphine, subject on both placebo relief: 31.1 in placebo:
extension study in Table mixed neuropathic methadone and 10 mg condition, 6 39.7 with dronabinol 10
2b) (NCT00153192) and nociceptive (N = hydrocodone, subjects during their mg: 41.7 with dronabinol
11), and hydromorphone) placebo condition only 20 mg
uncategorized (N = 5).
Mean age 43.3 years,
53% female
e. Observational studies
Aviram 2020 Prospective observational Adults with any form 12 months following Not specified, Varied products (74% NA 42% reduction [27 mg Not reported by opioid GRADE rating “low”
cohort study of chronic non- treatment initiation mixture of weak THC dominant). OMEDD (95% CI: −34.89, status large observational
cancer–related pain (n = 118) and Overall reported 18.56) reduction (p < cohort.
(n = 829, 66% strong (n = 56) monthly MC dose 0.001)). Opioid cessation In the same cohort,
retained at 12 opioids at increased from 20 ± at 12-month follow-up: of those not taking
months). Average age baseline 20–20 g at T1 to 30 ± 24% using weak and 20% opioids at baseline
47 years, 43% female 20–30 at T12 (χ2 (4) = using strong opioids; χ2 9% started weak
1250.32, p < 0.001). (5) = 27.3, p < 0.001; χ2 (5) opioids and 10%

Neuropsychopharmacology (2022) 47:1315 – 1330


= 21.9; p < 0.001, started strong
respectively opioids
Bellnier 2018 Observational pre-post Adults (n = 29) with 3 months Not specified, 10 mg capsules of 1:1 Pre-post- Opioid consumption was Paroxysmal pain GRADE rating “low”
study chronic pain (n = 3 calculated at THC and CBD taken reduced from 79.94 decreased from 6.76 to observational data
cancer-related, n = 26 OMEDD orally every 8–12 h. (range 0–450) to 19.65 2.04 (p < 0.0001); surface Quality of life EQ-
non-cancer pain, 20 Vapor pen inhaler of (range 0–150) morphine from 4.20 to 1.30 (p < 5D scores (range 0
with spinal cord injury THC/CBD (20:1, 2 mg equivalents per day (p < 0.0001); deep from 5.87 to 100) improved
with spasticity). THC per 0.1 mg CBD) 0.05). Most (n = 26) to 2.03 (p < 0.0001); and from 36.08 ± 19.85
Average age was 61 1 to 5 puffs every 15 completely discontinued unpleasant rating at baseline to 64.43
years (±10 years), and min until relief was opioid use, and the declined from “miserable” + 19.15 after 3
65% of the sample achieved and use remaining patients to “annoying” after 3 months treatment
were female every 4–6 h as reduced their doses by months therapy (p < 0.0001)
needed approximately 75%
Capano 2020 Prospective, Adults (n = 113) with 4 and 8 weeks Not stated, 15.7 mg CBD, 0.5 mg NA OMEDD not captured. Baseline pain (PEG) scale GRADE rating “low”
observational cohort mod-severe chronic requirement to THC, 0.3 mg Fifty of the 94 (53.2%) at (6.5 [95% CI: 6.16–6.81], observational data
study pain for at least 3 be taking at least cannabidivarin participants using the 4 weeks 5.9 [95% CI: with no control
years, and stable 50 mg OMEDD (CBDV), 0.9 mg CBD hemp extract were 5.55–6.25] and 8
opioids for at least 1 for 12 months cannabidiolic acid able to reduce opioid weeks,5.7 [95% CI:
year (n = 113, 74% prior to (CBDA), 0.8 mg medications at week 8. Of 5.31–6.12], p =0.006)
retained at 8 weeks, enrollment cannabichrome the fifty who reduced, (12% reduction in pain,
aged 30–65 years (CBC), and >1% two ceased completely [30% reduction
(mean 56.1 years), botanical terpene considered clinically
68% female blend significant])
Habib 2018 Retrospective cohort Adults aged 18 and Median cannabis Codeine, The mean dose of NA 4/4 patients on weak Not reported by opioid
S. Nielsen et al.

GRADE rating “very


study above with duration 3 months tramadol, medical cannabis opioids at baseline, status low”, small
fibromyalgia (n = 26), oxycodone, was 26 ± 8.3 g per ceased while taking retrospective
19 female patients fentanyl or month cannabinoids (p = 0.055); cohort with no
(73%), mean 37.8 ± 7.6 buprenorphine. 15/20 patients on strong control group, short
years Varied doses opioids ceased while follow-up
taking medical
cannabinoids (p = 0.000)
Haroutounian Prospective, Adults (n = 73/274) 18 6 months Morphine, Smoked (THC NA 32/73(44%) ceased Pain outcomes not GRADE rating “low”,
2016 observational cohort years + above 18 with oxycodone, 6–14%, CBD opioids, (p < 0.001). available for the non-randomized
study chronic pain, 73 fentanyl, 0.2–3.8%) and oral Median OMEDD among subsample on opioids single-arm open-
prescribed opioids. hydromorphone, (THC 11–19%, CBD participants receiving label study
Mean age 51.2 years, buprenorphine, 0.5–5.5%). The mean opioids at follow-up (n =
62% female methadone and (SD) monthly 41) decreased from 60
tramadol. prescribed amount mg (95% CI: 45–90) to 45
of cannabis was 43.2 mg (95% CI: 30-,90); (p =
(17.9) g (any 0.19, Mann–Whitney)
formulation)
Hickernell Retrospective cohort Adults (n = 243) who Mean length of stay Oral oxycodone Dronabinol 5 mg Patients who did No significant difference No significant difference GRADE rating “low”
2018 study had total knee or hip 2–3 days 10 mg up to three twice a day during not receive in OMEDD or total OME on pain scores between non-randomized
arthroplasty. Mean doses mg plus hospital stay (n = 81) dronabinol (n = dose/length of stay. the groups on any day retrospective study.
age 62.3 years, 64% immediate 162) over the Significantly lower total post-surgery Mean length of stay
female release same time OME consumption during lower for the
oxycodone 5–10 period stay: Dronabinol group dronabinol group
mg mg as 252.5 mg ± 131.5 mg, compared with
required control group 313.3 mg ± control (2.3 ± 0.9 vs
185.4 mg. p = 0088 3.0 ± 1.2 days, p =
0.02)
Hoggart 2015 Open-label extension Adults (n = 380) with 38 weeks Strong and weak THC/CBD oral NA No change in the GRADE rating
study from 2 clinical trials peripheral opioids mucosal spray (2.7 proportion of the whole “moderate”, non-
1323
Table 2. continued 1324
Study Study design Population Observation period Opioid used Cannabinoid Used Comparator Effect of cannabinoid on Outcome on analgesia GRADE evidence
reference opioid dose observed rating and other
notes
across 66 study sites (38 neuropathic pain, mg of THC and 2.5 sample taking strong randomized
centers in six countries) mean age 57.8 years, mg of CBD per spray) opioids (56/380 at Data on other outcomes sample. Rigorous
not provided by opioid
47% female max 24 sprays per baseline and 57/380 at use status data collection
day follow-up) or other
opioids (118/380 at
baseline to 123/380 at
follow-up) following
cannabinoid use
Lynch 2002 Observational case series Adults with pain 1–9 month Morphine (varied Smoked cannabis NA Mean baseline morphine Improved pain control GRADE rating “very
conditions (n = 3) observation period doses) plant, unknown dose 195 mg (SD 147 mg) described with patients low“, unblinded
(peripheral content compared with mean 35 either reducing or observational study
neuropathy, multiple mg (SD 31 mg) after ceasing morphine dose
sclerosis, lower back commencing smoked
pain), Aged 35–47 cannabis. Opioid dose
years, 33% female reduction or cessation in
each case
Maida 2008 Prospective observational Adults with advanced 30 days Nabilone group Nabilone, mean of People with Log OMEDD in nabilone Pain score in nabilone GRADE rating ”low”.
study cancer (n = 112), 47 of baseline OMEDD 1.79 mg daily advanced cancer group 3.8 mg compared group 3.7 compared with Nabilone
whom were treated 60.3 mg (SD 64.6); who were not with 4.3 mg in the 5.0 in the untreated prescribing based
with nabilone (mean comparison treated with untreated group (p = group (p = 0.003); on symptom-
age 67 years, 38% group OMEDD nabilone 0.016), remained remained significant after related distress on
female) 67.3 mg (SD significant after adjusting adjusting for baseline the initial
101.0) for baseline symptom symptom level and consultation,
level and propensity propensity score with leading to selection
score pain score of 3.0 in the bias, but managed
nabilone group and 5.5 in with propensity
the comparison group (p scoring
< 0.001)
Maida 2017 Observational case series Adults with pyoderma 6–25 days Opioid analgesic Topical cannabinoid NA Mean Baseline OMEDD Mean pain at baseline 8.6, GRADE rating “very
gangrenosum (n = 2) type not specified oil THC:CBD 5:6 mg/ 26.7 mg (SD 0.9), Mean mean pain at follow-up low”, very small case
on opioids. Female mL or THC:CBD 7:9 follow-up 6.4 mg (SD 8.7) was 2.6 (70% reduction, series
(50 years) and male mg/mL i.e., clinically meaningful
(76 years) reduction)
S. Nielsen et al.

Maida 2020 Observational case series Two adults (aged 86 57–68 days Case 1: Codeine Topical cannabinoid NA Both patients ceased Not reported, opioid GRADE rating “very
and 69, both female), (with product THC < 1 mg/ opioids requirements used as low”, very small case
with painful and non- acetaminophen), mL, CBD 3.75 mg/mL proxy for pain series
healing leg ulcers, of Case 2 188 mg
greater than 6 oral morphine
months duration equivalents
(opioid type not
stated)
Narang 2008 Open-label extension Patients on opioids Four weeks OMEDD mean Flexible dose NA Opioid dose not reported Mean baseline NRS of 6.9 GRADE rating “low”.
following randomized, for chronic pain; BPI ≥ 68.1 mg (SD 57.2, schedule; dronabinol compared with mean Improvements (p <
single-dose, double-blind, 4 (n = 28). (see Table range 7.5–228) 5 mg daily − 20 mg NRS of 5.2 after 4 weeks 0.05) in sleep,
crossover trial (Table 2d) 2d for participant (mix of three times a day. of dronabinol (24% energy, pain relief,
characteristics) oxycodone, reduction in pain). and social
morphine, Statistically significant functioning. Lack of
methadone reduction, but does not placebo control
hydrocodone, meet the 30% reduction means effects may
hydromorphone) in pain to be clinically be non-specific/
significant placebo
Rod 2019 Open-label prospective Patients with chronic Six months Mean OMEDD CBD and THC (4–6%). NA 156 patients (26%) Pain not quantified. One GRADE rating “low”,
opioid taper study pain (n = 600), on 120 mg (Range Doses related ceased opioids, and a patient increased opioid evidence-based
opioid doses of 90–240 mg) directly to the opioid further 329patients (55%) intake; all other patients online
90–240 mg (age and taper: 0.5 g/day for reduced opioid use by an expressed satisfaction psychological
gender not reported) each 10% reduction average of 30%. Cannabis with their pain control, support provided
in opioid dose, as use among these patients sleep and quality of life (e.g., cognitive
needed by ranged from 1–3 g/day behavioral therapy
sublingual, oral or and mindfulness)
inhalation by
vaporization
Safakish 2020 Prospective observational 82/751 chronic pain 12 months Mixed opioids, 7% to 29% THC and/ NA Baseline (n = 82) OMEDD Not reported by opioid GRADE rating “very
cohort study patients, who were converted to oral or CBD. 26.2 (SD 48.1), Month 1 status low”, open-label
using opioids. Mean morphine (N = 67) 12.1 (SD 44.7), single harm study
age of 49.6 years, 57% equivalent doses month 3 (N = 26) 3.3 (SD with high attrition
female 8.6), month 6 (n = 9) 3.0
(SD 6.5) month 12 (n = 4)
1.4 (SD 0.1). p < 0.001
Schneider- Retrospective matched Adults with traumatic 48–96 h after Not stated, opioid Dronabinol (usually Usual care OMEDD reduction in Adjunctive dronabinol GRADE rating “low”,
Smith 2020 cohort study injury: 33 cases (mean admission use reported in 5–10 mg twice a day) without group dronabinol (−79 reduce pain scores. non-randomized
age 39.9 years, 76% OMEDD dronabinol mg (SD20), p < 0.001), Average change in pain retrospective study
male) and 33 OMEDD for controls scores (NRS) were similar
matched controls unchanged from baseline between cases and
(mean age 30.0 years, (−9 (18) mg, p = 0.63) controls (−0.4 vs −0.6, p
30% female) = 0.78)

Neuropsychopharmacology (2022) 47:1315 – 1330


Takakuwa Retrospective cohort Adults with low back Data extracted from Variable products NA 31/61 ceased, 9 reduced Not reported GRADE rating “very
2020 study pain (n = 61) who 1997–2019 from a reported in grams and 15 increased their low”, small
S. Nielsen et al.
1325

GRADE Grading of Recommendations Assessment, Development and Evaluation, CBD cannabidiol, OME oral morphine equivalent, OMEDD oral morphine equivalent daily dose, IQR interquartile range, SD standard
arm study; selection
GRADE rating “very
nabiximols on change in OMEDD (Mean difference −3.8 mg, 95%

low”, small sample,

bias as only those


open-label single-
rating and other
GRADE evidence

standard therapy
retrospective
CI −10.97, 3.37, I2 = 23%) (Fig. 2a). Four studies (1109 participants)
cohort study

that did not


respond to
found no effect of nabiximols on percentage change in pain

included
scores (mean difference 1.84, 95% CI −2.05, 5.72, I2 = 58%)
notes

(Fig. 2b). Five studies (1536 participants) examined serious adverse


events and found no difference in events with cannabinoids
compared with placebo (risk ratio [RR] 1.23, 95% CI 0.89, 1.70, I2 =

0.0001. 60% reduction in


Outcome on analgesia

month: 3.3 (SD 2.2) p <


months: 5.3 (SD 1.3); 6
baseline 8.1(SD 1.4); 3
58%) (Fig. 2c). Five studies (1,536 participants) examined adverse

pain at 3 months, i.e.,


clinically meaningful
Mean VAS score at
events other than serious adverse events and found more non-
serious adverse events with cannabinoids compared with placebo
(RR 1.13, 95% CI 1.03, 1.24, I2 = 0%) (Fig. 2d).

reduction
observed

Clinical trials—chronic non-cancer pain. Five clinical trials (139


participants, Table 2d) examined the effects of dronabinol [73–75]
and smoked cannabis [76, 77] in patients with chronic non-cancer
Effect of cannabinoid on

24–6), sign test: p = 0.004,

90–5) intermittent opioid


Wilcoxon sign rank test: p

use (median = 5.8, IQR =


opioids. Median OMMED
reduction 9.0 mg (IQR =

pain. Most studies had short observation periods (5 h to 5 days)


analysis: chronic use
(median = 21, IQR =

[74–77], and used crossover designs [73–76]. Opioid dose was an


= 0.09). Subgroup

outcome in one study, with no difference between smoked


Not reported
opioid dose

cannabis and placebo [76]. All five studies reported on analgesic


outcomes with conflicting findings. A single-arm open-label study
12–6)

(with no comparison group) recruited people with mixed types of


chronic non-cancer pain (n = 24) who were prescribed opioids,
and found significant overall reductions from baseline pain ratings
Baseline status

following co-administration of cannabinoids [77]. In contrast, a


Comparator

on standard

double-blind crossover study in sickle cell patients found no


analgesic
therapy

significant differences analgesia effects between placebo and


vaporized cannabis [76]. Two studies recruited patients with
chronic pancreatitis and found no effect of dronabinol on pain
deviation, THC delta-9-tetrahydrocannabinol, NRS numerical rating scale, BPI Brief Pain Inventory, VAS visual analog scale.
vaporized) 1:4 THC to

option to increase to
grams per month for

measures compared with placebo [73, 74]. A sub-analysis in


cannabis use 1.45 g/

CBD. The THC levels

cannabinoid was 20
3 months, with the
Cannabinoid Used

were less than 5%.


0.01–18.7 g). Most

patients with chronic postsurgical abdominal pain found lower


per day. Median

smoked (44/61

pain among those who received dronabinol compared with


cannabinoids
day (range =

The dose of

30 g/month
(smoked or

thereafter

placebo [73]. A single-dose study in patients with mixed-chronic


Medical
(72%)).

pain conditions, found dronabinol 10 and 20 mg was associated


increased analgesia compared with placebo [75]. These studies
did not include measures of abuse liability.
Oxycodone 5 mg/
21 mg/day (range

analgesic therapy
Naloxone 2.5 mg
Median OMEDD

prior to trialing
cannabinoids)

Clinical studies—observational. Seventeen observational studies (n


(minimum 12
Opioid used

twice a day
= 1.1–500).

= 2674) examined the opioid-sparing effects of cannabinoids; three


months of
standard

medical

small retrospective case series of two to three patients each [78–80],


two retrospective cohort studies [81, 82], two retrospective matched
cohort studies [83, 84], and ten prospective observational cohort
studies [85–93], including two open-label extension studies [75, 93]
Observation period

than 3000 patients


database of more

(see Table 2e). Two retrospective matched cohort studies examined


acute analgesia with traumatic injury [83] and joint arthroplasty [84].
Both found no difference in pain scores, but reduced opioid
6 months

consumption on at least one measure. For pain management


following joint arthroplasty, there was no change in daily opioid dose
Data extracted from publication in addition to clinialtrials.gov.

with dronabinol administration, but a reduced total opioid


consumption due to significantly shorter hospital stays in the
duloxetine. Mean age
fibromyalgia already
50.1 years, (range =
49–86), 38% female

Adult (n = 31) with


low back pain and
opioids. Mean age

dronabinol group [84]. One study compared those prescribed


were prescribed

33.5 years., 90%


oxycodone and

nabilone with those that had not received it, using propensity
treated with
Population

scoring to adjust for the greater severity of the nabilone prescribed


female

group [89]. The remaining observational studies did not have control
conditions and examined opioid use in patients with a range of
different types of chronic non-cancer pain. Seven studies reported on
the outcome of OMEDD after commencing medical cannabinoids,
Prospective observational

with reductions from 9 to 140 mg OMEDD reported (Table 2b). Four


studies quantified the reduction in pain scores, which ranged from
crossover study

12% to 70%, with two studies exceeding the minimum threshold of a


Study design

30% reduction in pain to be clinically meaningful. Meta-analysis was


possible for studies that reported the proportion of patients who
continued

reported opioid reduction or cessation; eight studies reported the


proportion of patients who ceased opioids (range 2–100%), with a
pooled prevalence of 0.39 (95% CI 0.15, 0.64, I2 = 95.47%)
(Appendix 5a). Seven studies reported on the proportion of patients
Yassin 2019
reference
Table 2.

reducing opioid use (range 44–100%) with a pooled prevalence of


Study

0.85 (95% CI 0.64, 0.99, I2 = 92.82%) (Appendix 5b). Statistically


a

significant heterogeneity was identified in both meta-analyses.

Neuropsychopharmacology (2022) 47:1315 – 1330


S. Nielsen et al.
1326

Fig. 1 Forrest plot for meta-analysis examining the opioid-sparing effect of delta-9-THC when co-administered with morphine. Note
mean difference and standard deviation values are of log10ED50.

(a)

(b)

(c)

(d)

Fig. 2 Opioid-sparing outcomes from clinical trials in people with cancer pain. Meta-analysis comparing cannabinoids with placebo on
outcomes of a percent improvement in pain score, b change in mean total Oral Morphine Equivalent Daily Dose (OMEDD), c serious adverse
events from baseline, and d adverse events excluding serious adverse events, in clinical trials of people with cancer pain.

Neuropsychopharmacology (2022) 47:1315 – 1330


S. Nielsen et al.
1327
Quality ratings of clinical studies Finally, observational studies had methodological concerns
The clinical studies were rated using the GRADE criteria. Of the including small sample sizes (several observational studies
clinical trials, five laboratory studies provided moderate evidence, included in meta-analysis had two to three patients), no control
three clinical trials in acute pain provided high quality evidence, groups or blinding, selection bias, and were likely to have been
six clinical studies provided low-high quality evidence in cancer impacted by expectancy effects.
pain, and five studies in chronic non-cancer pain were assessed as Although our review is much broader, we have drawn similar
low-moderate quality. The seventeen observational studies were conclusions to earlier reviews. For example, a review of cross-
assessed to be low to very-low-quality evidence (Table 2). sectional surveys and cohort studies, representing lower quality
evidence, found large reductions in opioid doses, though study
Ongoing clinical trials. We identified 15 registered clinical trials designs prevented the drawing of causal conclusions [95]. A later
which, based on published protocols and clinical trial registry review with five randomized trials with patients with chronic pain
entries, may provide important data for future updated reviews and 12 observational studies further concluded that there was
(Appendix 6). uncertainty in the evidence [96], although this review considered
a substantially smaller number of clinical trials than we consider.
Future studies may benefit from focusing on populations with
DISCUSSION higher opioid tolerance, or higher motivation to reduce opioid
The current update represents the largest synthesis of studies doses, where clinical benefits may be greatest [97]. Standardiza-
examining the opioid-sparing effects of cannabinoids, with double tion of outcomes for opioid-sparing research may assist with
the number of preclinical studies, four times as many clinical harmonization of outcome measures and support meta-analysis
studies and more than six times the number of participants with future clinical trials [2].
(>5000) compared to our earlier review [3], reflecting the rapid Despite the inclusion of a larger number of studies, and the
growth of clinical research in this area. increased size and quality of clinical trials in recent years, our
Most preclinical studies found synergistic effects with opioids and conclusions have not changed substantially from our earlier
cannabinoids co-administration, predominantly with mixed CB1/CB2 review. Nevertheless, we did identify 15 registered clinical trials
agonists such as delta-9-THC, though effects varied with different indicating that this continues to be an active area of research in
cannabinoids, opioids and pain assays. Meta-analyses (with one which the science is likely to continue to evolve.
addition preclinical study since 2015) demonstrated that morphine In conclusion, preclinical studies support the opioid-sparing
dose required to produce an equivalent analgesic effect was 3.5 effect of delta-9-THC and other mixed CB1/CB2 agonists.
times lower when co-administered with delta-9-THC, consistent with Observational studies support the opioid-sparing potential of
the previous review [3]. This effect would be clinically meaningful if cannabinoids. However, findings from clinical trials provide
replicated in well-controlled clinical studies. However, preclinical conflicting results that may highlight important areas for future
studies often have larger effect sizes, attributed to the reduced research. These include identifying optimal doses and popula-
heterogeneity compared to clinical populations [94]. This body of tions who may experience benefits with cannabinoids. With
preclinical research may help to identify specific cannabinoids and numerous clinical trials currently underway, we will update our
mechanisms that underlie an opioid-sparing effect, with the most review, as higher-quality data may enable stronger conclusions
consistent effects observed with mixed CB1/CB2 agonists, and to be made.
evidence of potential antagonistic effects between CB1 agonist and
mu receptor agonists in models of mechanical hyperalgesia.
A rapidly growing number of clinical studies have measured REFERENCES
opioid-sparing endpoints, though findings were inconsistent. The 1. Dowell D, Haegerich TM, Chou R. CDC guideline for prescribing opioids for
highest quality studies were conducted in patients with cancer chronic pain—United States, 2016. JAMA. 2016;315:1624–45.
pain, where meta-analysis of four studies did not find significant 2. Gewandter JS, Smith SM, Dworkin RH, Turk DC, Gan TJ, Gilron I, et al. Research
approaches for evaluating opioid sparing in clinical trials of acute and chronic
effects on opioid dose or analgesia. Conflicting findings were
pain treatments: Initiative on Methods, Measurement, and Pain Assessment in
found in studies of experimental pain, and in patients with chronic Clinical Trials recommendations. Pain. 2021;162:2669–81.
non-cancer pain. Further studies are needed to clarify the results 3. Nielsen S, Sabioni P, Trigo JM, Ware MA, Betz-Stablein BD, Murnion B, et al.
found here given the small number of studies. Opioid-sparing effect of cannabinoids: a systematic review and meta-analysis.
A limited number of controlled studies demonstrated benefits Neuropsychopharmacology. 2017;42:1752–65.
of combining cannabinoids with opioids for analgesia. Experi- 4. Campbell G, Hall W, Nielsen S. What does the ecological and epidemiological
mental pain studies found cannabinoids improved [62, 63] and evidence indicate about the potential for cannabinoids to reduce opioid use and
worsened [61] analgesia. These effects were not dose dependent, harms? A comprehensive review. Int Rev Psychiatry. 2018;30:91–106.
with significant effects seen with lower but not higher doses of 5. Shover CL, Davis CS, Gordon SC, Humphreys K. Association between medical
cannabis laws and opioid overdose mortality has reversed over time. Proc Natl
delta-9-THC. Opioid-sparing effects were not seen in well-
Acad Sci USA. 2019;116:12624.
conducted RCTs with acute pain, or in meta-analyses of RCTs in 6. Desroches J, Beaulieu P. Opioids and cannabinoids interactions: involvement in
cancer pain, and studies that did find positive effects have pain management. Curr Drug Targets. 2010;11:462–73.
important limitations such as no control group [77], small sample 7. Hojo M, Sudo Y, Ando Y, Minami K, Takada M, Matsubara T, et al. mu-Opioid
sizes [67, 75], and the mixed quality of the study design. receptor forms a functional heterodimer with cannabinoid CB1 receptor: elec-
Furthermore, some RCTs instructed patients to continue their trophysiological and FRET assay analysis. J Pharm Sci. 2008;108:308–19.
pain medication in the same doses, which may preclude 8. Babalonis S, Walsh SL. Therapeutic potential of opioid/cannabinoid combinations
identifying a change in opioid dose [70–73, 77], although changes in humans: review of the evidence. Eur Neuropsychopharmacol. 2020;36:206–16.
in breakthrough opioid requirements were a secondary outcome 9. An D, Peigneur S, Hendrickx LA, Tytgat J. Targeting cannabinoid receptors: cur-
rent status and prospects of natural products. Int J Mol Sci. 2020;21:5064.
in six studies [69–72, 75]. Some clinical studies demonstrated
10. Yao BB, Mukherjee S, Fan Y, Garrison TR, Daza AV, Grayson GK, et al. In vitro
beneficial effects of opioid and cannabinoid co-administration on pharmacological characterization of AM1241: a protean agonist at the cannabi-
other outcomes such as sleep, and functioning in chronic pain noid CB2 receptor? Br J Pharmacol. 2006;149:145–54.
patients [75, 77]. Conflicting results were found between 11. Lu H-C, Mackie K. An introduction to the endogenous cannabinoid system. Biol
preclinical studies and clinical trials on measure of abuse liability. Psychiatry. 2016;79:516–25.
Evidence of reduced abuse liability was found in some animal 12. Moher D, Liberati A, Tetzlaff J, Altman DG. Preferred reporting items for sys-
models, which contrasted directly with evidence of increased drug tematic reviews and meta-analyses: the PRISMA statement. PLoS Med. 2009;6:
liking and subjective effects in human studies. e1000097.

Neuropsychopharmacology (2022) 47:1315 – 1330


S. Nielsen et al.
1328
13. Covidence systematic review software. covidence.org. Veritas Health Innovation: 37. Williams IJ, Edwards S, Rubo A, Haller VL, Stevens DL, Welch SP. Time course of
Melbourne, Australia; 2019. the enhancement and restoration of the analgesic efficacy of codeine and
14. Litchfield JA, Wilcoxon F. A simplified method of evaluating dose-effect experi- morphine by delta9-tetrahydrocannabinol. Eur J Pharmacol. 2006;539:57–63.
ments. J Pharmacol Exp Ther. 1949;96:99–113. 38. Alsalem M, Altarifi A, Haddad M, Aldossary SA, Kalbouneh H, Aldaoud N, et al.
15. Luo D, Wan X, Liu J, Tong T. Optimally estimating the sample mean from the Antinociceptive and abuse potential effects of cannabinoid/opioid combinations
sample size, median, mid-range, and/or mid-quartile range. Stat Methods Med in a chronic pain model in rats. Brain Sci. 2019;9:328.
Res. 2016;27:1785–805. 39. Alsalem M, Altarifi A, Haddad M, Azab B, Kalbouneh H, Imraish A, et al. Analgesic
16. Wan X, Wang W, Liu J, Tong T. Estimating the sample mean and standard effects and impairment in locomotor activity induced by cannabinoid/opioid
deviation from the sample size, median, range and/or interquartile range. BMC combinations in rat models of chronic pain. Brain Sci. 2020;10:1–18.
Med Res Methodol. 2014;14:135. 40. Altun A, Yildirim K, Ozdemir E, Bagcivan I, Gursoy S, Durmus N. Attenuation of
17. Guyatt GH, Oxman AD, Vist GE, Kunz R, Falck-Ytter Y, Alonso-Coello P, et al. morphine antinociceptive tolerance by cannabinoid CB1 and CB2 receptor
GRADE: an emerging consensus on rating quality of evidence and strength of antagonists. J Physiological Sci. 2015;65:407–15.
recommendations. Br Med J. 2008;336:924–6. 41. Auh QS, Chun YH, Melemedjian OK, Zhang Y, Ro JY. Peripheral interactions
18. Sterne JAC, Sutton AJ, Ioannidis JPA, Terrin N, Jones DR, Lau J, et al. Recom- between cannabinoid and opioid receptor agonists in a model of inflammatory
mendations for examining and interpreting funnel plot asymmetry in meta- mechanical hyperalgesia. Brain Res Bull. 2016;125:211–17.
analyses of randomised controlled trials. BMJ. 2011;343:d4002. 42. Chen X, Cowan A, Inan S, Geller EB, Meissler JJ, Rawls SM, et al. Opioid-sparing
19. Cichewicz DL, Martin ZL, Smith FL, Welch SP. Enhancement mu opioid anti- effects of cannabinoids on morphine analgesia: participation of CB1 and CB2
nociception by oral delta9-tetrahydrocannabinol: dose-response analysis and receptors. Br J Pharmacol. 2019;176:3378–89.
receptor identification. J Pharmacol Exp Ther. 1999;289:859–67. 43. Grenald SA, Young MA, Wang Y, Ossipov MH, Ibrahim MM, Largent-Milnes TM,
20. Cichewicz DL, McCarthy EA. Antinociceptive synergy between delta(9)-tetra- et al. Synergistic attenuation of chronic pain using mu opioid and cannabinoid
hydrocannabinol and opioids after oral administration. J Pharmacol Exp Ther. receptor 2 agonists. Neuropharmacology. 2017;116:59–70.
2003;304:1010–5. 44. Iyer V, Slivicki RA, Thomaz AC, Crystal JD, Mackie K, Hohmann AG. The canna-
21. Cox ML, Haller VL, Welch SP. Synergy between delta9-tetrahydrocannabinol and binoid CB2 receptor agonist LY2828360 synergizes with morphine to suppress
morphine in the arthritic rat. Eur J Pharmacol. 2007;567:125–30. neuropathic nociception and attenuates morphine reward and physical depen-
22. Smith FL, Cichewicz D, Martin ZL, Welch SP. The enhancement of morphine dence. Eur J Pharmacol. 2020;886:173544.
antinociception in mice by delta9-tetrahydrocannabinol. Pharmacology Biochem 45. Kazantzis NP, Casey SL, Seow PW, Mitchell VA, Vaughan CW. Opioid and can-
Behav. 1998;60:559–66. nabinoid synergy in a mouse neuropathic pain model. Br J Pharmacol.
23. Williams J, Haller VL, Stevens DL, Welch SP. Decreased basal endogenous opioid 2016;173:2521–31.
levels in diabetic rodents: effects on morphine and delta-9-tetrahydrocannabinoid- 46. Maguire DR, France CP. Additive antinociceptive effects of mixtures of the kappa-
induced antinociception. Eur J Pharmacol. 2008;584:78–86. opioid receptor agonist spiradoline and the cannabinoid receptor agonist
24. Welch SP, Stevens DL. Antinociceptive activity of intrathecally administered CP55940 in rats. Behavioural Pharmacol. 2016;27:69–72.
cannabinoids alone, and in combination with morphine, in mice. J Pharmacol Exp 47. Maguire DR, France CP. Antinociceptive effects of mixtures of mu opioid receptor
Ther. 1992;262:10–8. agonists and cannabinoid receptor agonists in rats: impact of drug and fixed-
25. Wakley AA, Craft RM. THC-methadone and THC-naltrexone interactions on dis- dose ratio. Eur J Pharmacol. 2018;819:217–24.
crimination, antinociception, and locomotion in rats. Behavioural Pharmacol. 48. Minervini V, Dahal S, France CP. Behavioral characterization of κ opioid receptor
2011;22:489–97. agonist spiradoline and cannabinoid receptor agonist CP55940 mixtures in rats. J
26. Wilson AR, Maher L, Morgan MM. Repeated cannabinoid injections into the rat Pharmacol Exp Ther. 2017;360:280–87.
periaqueductal gray enhance subsequent morphine antinociception. Neuro- 49. Neelakantan H, Tallarida RJ, Reichenbach ZW, Tuma RF, Ward SJ, Walker EA.
pharmacology. 2008;55:1219–25. Distinct interactions of cannabidiol and morphine in three nociceptive behavioral
27. Yesilyurt O, Dogrul A, Gul H, Seyrek M, Kusmez O, Ozkan Y, et al. Topical can- models in mice. Behavioural Pharmacol. 2015;26:304–14.
nabinoid enhances topical morphine antinociception. Pain. 2003;105:303–8. 50. Nguyen JD, Grant Y, Creehan KM, Hwang CS, Vandewater SA, Janda KD, et al. Δ 9
28. Tham SM, Angus JA, Tudor EM, Wright CE. Synergistic and additive interactions of -tetrahydrocannabinol attenuates oxycodone self-administration under extended
the cannabinoid agonist CP55,940 with mu opioid receptor and alpha2- access conditions. Neuropharmacology. 2019;151:127–35.
adrenoceptor agonists in acute pain models in mice. Br J Pharmacol. 51. Nilges MR, Bondy ZB, Grace JA, Winsauer PJ. Opioid-enhancing antinociceptive
2005;144:875–84. effects of delta-9-tetrahydrocannabinol and amitriptyline in rhesus macaques.
29. Smith PA, Selley DE, Sim-Selley LJ, Welch SP. Low dose combination of morphine Exp Clin Psychopharmacol. 2020;28:355–64.
and delta9-tetrahydrocannabinol circumvents antinociceptive tolerance and 52. Rodríguez-Muñoz M, Onetti Y, Cortés-Montero E, Garzón J, Sánchez-Blázquez P.
apparent desensitization of receptors. Eur J Pharmacol. 2007;571:129–37. Cannabidiol enhances morphine antinociception, diminishes NMDA-mediated
30. Reche I, Fuentes JA, Ruiz-Gayo M. Potentiation of delta 9-tetrahydrocannabinol- seizures and reduces stroke damage via the sigma 1 receptor. Mol Brain.
induced analgesia by morphine in mice: involvement of mu- and kappa-opioid 2018;11:51.
receptors. Eur J Pharmacol. 1996;318:11–6. 53. Sierra S, Gupta A, Gomes I, Fowkes M, Ram A, Bobeck EN, et al. Targeting can-
31. Pugh G Jr, Smith PB, Dombrowski DS, Welch SP. The role of endogenous opioids nabinoid 1 and delta opioid receptor heteromers alleviates chemotherapy-
in enhancing the antinociception produced by the combination of DELTA9- induced neuropathic pain. ACS Pharmacol Transl Sci. 2019;2:219–29.
tetrahydrocannabinol and morphine in the spinal cord. J Pharmacol Exp Ther. 54. Yuill MB, Hale DE, Guindon J, Morgan DJ. Anti-nociceptive interactions between
1996;279:608–16. opioids and a cannabinoid receptor 2 agonist in inflammatory pain. Mol Pain.
32. Maguire DR, Yang W, France CP. Interactions between mu-opioid receptor ago- 2017;13(no pagination).
nists and cannabinoid receptor agonists in rhesus monkeys: antinociception, 55. Zhang M, Chi M, Zou H, Tian S, Zhang Z, Wang G. Effects of coadministration of low
drug discrimination, and drug self-administration. [Erratum appears in J Phar- dose cannabinoid type 2 receptor agonist and morphine on vanilloid receptor 1
macol Exp Ther. 2014 Mar;348(3):490-1 Note: Dosage error in article text]. J expression in a rat model of cancer pain. Mol Med Rep. 2017;16:7025–31.
Pharmacol Exp Ther. 2013;345:354–62. 56. Zhang M, Dong L, Zou H, Li J, Li Q, Wang G, et al. Effects of cannabinoid type 2
33. Li JX, McMahon LR, Gerak LR, Becker GL, France CP. Interactions between receptor agonist AM1241 on morphine-induced antinociception, acute and
Delta(9)-tetrahydrocannabinol and mu opioid receptor agonists in rhesus chronic tolerance, and dependence in mice. J Pain. 2018;19:1113–29.
monkeys: discrimination and antinociception. Psychopharmacology. 2008;199: 57. Zhang M, Wang K, Ma M, Tian S, Wei N, Wang G. Low-dose cannabinoid type 2
199–208. receptor agonist attenuates tolerance to repeated morphine administration via
34. Katsuyama S, Mizoguchi H, Kuwahata H, Komatsu T, Nagaoka K, Nakamura H, regulating mu-opioid receptor expression in walker 256 tumor-bearing rats.
et al. Involvement of peripheral cannabinoid and opioid receptors in beta- Anesthesia Analgesia. 2016;122:1031–37.
caryophyllene-induced antinociception. Eur J Pain. 2013;17:664–75. 58. Stachtari CC, Thomareis ON, Tsaousi GG, Karakoulas KA, Chatzimanoli FI, Chat-
35. Finn DP, Beckett SR, Roe CH, Madjd A, Fone KC, Kendall DA, et al. Effects of zopoulos SA, et al. Interaction of a cannabinoid-2 agonist with tramadol on
coadministration of cannabinoids and morphine on nociceptive behaviour, brain nociceptive thresholds and immune responses in a rat model of incisional pain.
monoamines and HPA axis activity in a rat model of persistent pain. Eur J Neu- Am J Therapeutics. 2016;23:e1484–e92.
rosci. 2004;19:678–86. 59. Naef M, Curatolo M, Petersen-Felix S, Arendt-Nielsen L, Zbinden A, Brenneisen R.
36. Cichewicz DL, Welch SP, Smith FL. Enhancement of transdermal fentanyl and The analgesic effect of oral delta-9-tetrahydrocannabinol (THC), morphine, and a
buprenorphine antinociception by transdermal delta9-tetrahydrocannabinol. Eur THC-morphine combination in healthy subjects under experimental pain condi-
J Pharmacol. 2005;525:74–82. tions. Pain. 2003;105:79–88.

Neuropsychopharmacology (2022) 47:1315 – 1330


S. Nielsen et al.
1329
60. Roberts JD, Gennings C, Shih M. Synergistic affective analgesic interaction 82. Takakuwa KM, Hergenrather JY, Shofer FS, Schears RM. The impact of medical
between delta-9- tetrahydrocannabinol and morphine. Eur J Pharmacol. cannabis on intermittent and chronic opioid users with back pain: how cannabis
2006;530:54–58. diminished prescription opioid usage. Cannabis Cannabinoid Res. 2020;5:263–70.
61. Babalonis S, Lofwall MR, Sloan PA, Nuzzo PA, Fanucchi LC, Walsh SL. Cannabinoid 83. Schneider-Smith E, Salottolo K, Swartwood C, Melvin C, Madayag RM, Bar-Or D.
modulation of opioid analgesia and subjective drug effects in healthy humans. Matched pilot study examining cannabis-based dronabinol for acute pain fol-
Psychopharmacology. 2019;236:3341–52. lowing traumatic injury. Trauma Surg Acute Care Open. 2020;5:e000391.
62. Dunn KE, Bergeria CL, Huhn AS, Speed TJ, Mun CJ, Vandrey R, et al. Within- 84. Hickernell TR, Lakra A, Berg A, Cooper HJ, Geller JA, Shah RP. Should cannabinoids
subject, double-blinded, randomized, and placebo-controlled evaluation of the be added to multimodal pain regimens after total hip and knee arthroplasty? J
combined effects of the cannabinoid dronabinol and the opioid hydromorphone Arthroplast. 2018;33:3637–41.
in a human laboratory pain model. Neuropsychopharmacology. 2021;46:1451–59. 85. Aviram J, Pud D, Gershoni T, Schiff-Keren B, Ogintz M, Vulfsons S, et al. Medical
63. Cooper ZD, Bedi G, Ramesh D, Balter R, Comer SD, Haney M. Impact of co- cannabis treatment for chronic pain: outcomes and prediction of response. Eur J
administration of oxycodone and smoked cannabis on analgesia and abuse lia- Pain. 2021;25:359–74.
bility. Neuropsychopharmacology. 2018;43:2046–55. 86. Capano A, Weaver R, Burkman E. Evaluation of the effects of CBD hemp extract
64. Seeling W, Kneer L, Buchele B, Gschwend J, Maier L, Nett C, et al. DELTA9- on opioid use and quality of life indicators in chronic pain patients: a prospective
tetrahydrocannabinol and the opioid receptor agonist piritramide do not act cohort study. Postgrad Med. 2020;132:56–61.
synergistically in postoperative pain. [German] Keine synergistische wirkung der 87. Haroutounian S, Ratz Y, Ginosar Y, Furmanov K, Saifi F, Meidan R, et al. The effect
kombination von DELTA9-tetrahydrocannabinol und piritramid bei post- of medicinal cannabis on pain and quality-of-life outcomes in chronic pain: a
operativen schmerzen. Anaesthesist. 2006;55:391–400. prospective open-label study. Clin J Pain. 2016;32:1036–43.
65. Levin DN, Dulberg Z, Chan AW, Hare GM, Mazer CD, Hong A. A randomized- 88. Safakish R, Ko G, Salimpour V, Hendin B, Sohanpal I, Loheswaran G, et al. Medical
controlled trial of nabilone for the prevention of acute postoperative nausea and cannabis for the management of pain and quality of life in chronic pain patients:
vomiting in elective surgery. Can J Anaesth. 2017;64:385–95. a prospective observational study. Pain Med. 2020;18:3073–86.
66. Bebee B, Taylor DM, Bourke E, Pollack K, Foster L, Ching M, et al. The CANBACK 89. Maida V, Ennis M, Irani S, Corbo M, Dolzhykov M. Adjunctive nabilone in cancer
trial: a randomised, controlled clinical trial of oral cannabidiol for people pre- pain and symptom management: a prospective observational study using pro-
senting to the emergency department with acute low back pain. Med J Aust. pensity scoring. J Support Oncol. 2008;6:119–24.
2021;214:370–75. 90. Bellnier T, Brown GW, Ortega TR. Preliminary evaluation of the efficacy, safety,
67. Lissoni P, Porro G, Messina G, Porta E, Rovelli F, Roselli MG, et al. Morphine, and costs associated with the treatment of chronic pain with medical cannabis.
melatonin, Marijuana, Magnolia and MYRRH as the “five m” schedule in the Ment Health Clin. 2018;8:110–15.
treatment of cancer pain and the possible dose-dependency of the antitumor 91. Rod K. A pilot study of a medical cannabis – opioid reduction program. Am J
and analgesic effects of the pineal hormone melatonin. Anticancer Res. Psychiatry Neurosci. 2019;7:74–7.
2014;34:6033–34. 92. Yassin M, Oron A, Robinson D. Effect of adding medical cannabis to analgesic
68. Zylla DM, Eklund J, Gilmore G, Gavenda A, Guggisberg J, VazquezBenitez G, et al. treatment in patients with low back pain related to fibromyalgia: an observa-
A randomized trial of medical cannabis in patients with stage IV cancers to assess tional cross-over single centre study. Clin Exp Rheumatol. 2019;37(Suppl
feasibility, dose requirements, impact on pain and opioid use, safety, and overall 116):13–20.
patient satisfaction. Supportive Care Cancer. 2021;29:7471–78. 93. Hoggart B, Ratcliffe S, Ehler E, Simpson KH, Hovorka J, Lejčko J, et al. A multi-
69. Johnson JR, Burnell-Nugent M, Lossignol D, Ganae-Motan ED, Potts R, Fallon MT. centre, open-label, follow-on study to assess the long-term maintenance of
Multicenter, double-blind, randomized, placebo-controlled, parallel-group study effect, tolerance and safety of THC/CBD oromucosal spray in the management of
of the efficacy, safety, and tolerability of THC:CBD extract and THC extract in neuropathic pain. J Neurol. 2015;262:27–40.
patients with intractable cancer-related pain. J Pain Symptom Manag. 94. Berge O-G. Predictive validity of behavioural animal models for chronic pain. Br J
2010;39:167–79. Pharmacol. 2011;164:1195–206.
70. Portenoy RK, Ganae-Motan ED, Allende S, Yanagihara R, Shaiova L, Weinstein S, 95. Okusanya BO, Asaolu IO, Ehiri JE, Kimaru LJ, Okechukwu A, Rosales C. Medical
et al. Nabiximols for opioid-treated cancer patients with poorly-controlled cannabis for the reduction of opioid dosage in the treatment of non-cancer
chronic pain: a randomized, placebo-controlled, graded-dose trial. J Pain. chronic pain: a systematic review. Syst Rev. 2020;9:167.
2012;13:438–49. 96. Noori A, Miroshnychenko A, Shergill Y, Ashoorion V, Rehman Y, Couban RJ, et al.
71. Fallon MT, Albert Lux E, McQuade R, Rossetti S, Sanchez R, Sun W, et al. Sativex Opioid-sparing effects of medical cannabis or cannabinoids for chronic pain: a
oromucosal spray as adjunctive therapy in advanced cancer patients with chronic systematic review and meta-analysis of randomised and observational studies.
pain unalleviated by optimized opioid therapy: two double-blind, randomized, BMJ Open. 2021;11:e047717.
placebo-controlled phase 3 studies. Br J Pain. 2017;11:119–33. 97. Le Foll B. Opioid-sparing effects of cannabinoids: myth or reality? Prog Neu-
72. Lichtman AH, Lux EA, McQuade R, Rossetti S, Sanchez R, Sun W, et al. Results of a ropsychopharmacol Biol Psychiatry. 2021;106:110065.
double-blind, randomized, placebo-controlled study of nabiximols oromucosal
spray as an adjunctive therapy in advanced cancer patients with chronic
uncontrolled pain. J Pain Symptom Manag. 2018;55:179–88.e1.
AUTHOR CONTRIBUTIONS
73. de Vries M, van Rijckevorsel DCM, Vissers KCP, Wilder-Smith OHG, van Goor H.
SN, BM, NL, K-EK, MF, and BLF were involved in the conceptualization of the work. SN,
Tetrahydrocannabinol does not reduce pain in patients with chronic abdominal
LMP, BW, BM, JM, GC, MG, LP, and K-EK screened the abstracts and full texts. SN, LMP,
pain in a phase 2 placebo-controlled study. Clin Gastroenterol Hepatol.
BW, JM, GC, LP, MG, and BM extracted the data, and/or checked the extracted data.
2017;15:1079–86.e4.
BB-S transformed the preclinical data, prepared the data for meta-analysis and
74. De Vries M, Van Rijckevorsel DCM, Vissers KCP, Wilder-Smith OHG, Van Goor H.
provided advice on the meta-analysis. SN conducted the meta-analysis and drafted
Single dose delta-9-tetrahydrocannabinol in chronic pancreatitis patients: analgesic
the manuscript with assistance from BM, BW, LMP, LP, and K-EK. All authors revised
efficacy, pharmacokinetics and tolerability. Br J Clin Pharmacol. 2016;81:525–37.
the manuscript and approved the final version.
75. Narang S, Gibson D, Wasan AD, Ross EL, Michna E, Nedeljkovic SS, et al. Efficacy of
dronabinol as an adjuvant treatment for chronic pain patients on opioid therapy.
J Pain. 2008;9:254–64.
76. Abrams DI, Couey P, Dixit N, Sagi V, Hagar W, Vichinsky E, et al. Effect of inhaled FUNDING
cannabis for pain in adults with sickle cell disease: a randomized clinical trial. SN is supported by a NHMRC Research Fellowship (#1163961). Open Access funding
JAMA Network Open. 2020;3:e2010874. enabled and organized by CAUL and its Member Institutions.
77. Abrams DI, Couey P, Shade SB, Kelly ME, Benowitz NL. Cannabinoid-opioid
interaction in chronic pain. Clin Pharmacol Therapeutics. 2011;90:844–51.
78. Lynch ME, Clark AJ. Cannabis reduces opioid dose in the treatment of chronic
COMPETING INTERESTS
non-cancer pain. J Pain Symptom Manag. 2003;25:496–8.
SN, NL and MF have been investigators on an untied education grant from
79. Maida V, Corban J. Topical medical cannabis: a new treatment for wound pain—
Indivior for unrelated work. SN has received untied research grants from Seqirus to
three cases of pyoderma gangrenosum. J Pain Symptom Manag. 2017;54:732–36.
study prescription opioid overdose. BLF has received cannabis product from Aurora,
80. Maida V, Shi RB, Fazzari FGT, Zomparelli L. Topical cannabis-based medicines – a
Grants from Canopy and Alkermes, participated in an Advisory Board for Indivior (and
novel paradigm and treatment for non-uremic calciphylaxis leg ulcers: an open
few other industry related projects not related to the present work). NL has served on
label trial. Int Wound J. 2020;17:1508–16.
Advisory Boards for Chiesi Pharmaceuticals and Indivior, received research funding
81. Habib G, Artul S. Medical cannabis for the treatment of fibromyalgia. J Clin
from Camarus AB. K-EK has previously received a speaker’s honorarium from Pfizer
Rheumatol. 2018;24:255–58.
and Mundipharma, in addition to fees from an advisory board and an educational

Neuropsychopharmacology (2022) 47:1315 – 1330


S. Nielsen et al.
1330
grant from Seqirus. MG is funded through the New South Wales Health Clinical Open Access This article is licensed under a Creative Commons
Cannabis Medicines Program. All other authors have nothing to declare. Attribution 4.0 International License, which permits use, sharing,
adaptation, distribution and reproduction in any medium or format, as long as you give
appropriate credit to the original author(s) and the source, provide a link to the Creative
ADDITIONAL INFORMATION Commons license, and indicate if changes were made. The images or other third party
Supplementary information The online version contains supplementary material material in this article are included in the article’s Creative Commons license, unless
available at https://doi.org/10.1038/s41386-022-01322-4. indicated otherwise in a credit line to the material. If material is not included in the
article’s Creative Commons license and your intended use is not permitted by statutory
Correspondence and requests for materials should be addressed to Suzanne Nielsen. regulation or exceeds the permitted use, you will need to obtain permission directly
from the copyright holder. To view a copy of this license, visit http://creativecommons.
Reprints and permission information is available at http://www.nature.com/reprints org/licenses/by/4.0/.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims
© The Author(s) 2022
in published maps and institutional affiliations.

Neuropsychopharmacology (2022) 47:1315 – 1330

You might also like