Abstract

Objective

To evaluate the short-term and long-term effects of plant-based medical cannabis in a chronic pain population over the course of one year.

Design

A longitudinal, prospective, 12-month observational study.

Setting

Patients were recruited and treated at a clinic specializing in medical cannabis care from October 2015 to March 2019.

Subjects

A total of 751 chronic pain patients initiating medical cannabis treatment.

Methods

Study participants completed the Brief Pain Inventory and the 12-item Short Form Survey (SF-12), as well as surveys on opioid medication use and adverse events, at baseline and once a month for 12 months.

Results

Medical cannabis treatment was associated with improvements in pain severity and interference (P < 0.001) observed at one month and maintained over the 12-month observation period. Significant improvements were also observed in the SF-12 physical and mental health domains (P < 0.002) starting at three months. Significant decreases in headaches, fatigue, anxiety, and nausea were observed after initiation of treatment (P ≤ 0.002). In patients who reported opioid medication use at baseline, there were significant reductions in oral morphine equivalent doses (P < 0.0001), while correlates of pain were significantly improved by the end of the study observation period.

Conclusions

Taken together, the findings of this study add to the cumulative evidence in support of plant-based medical cannabis as a safe and effective treatment option and potential opioid medication substitute or augmentation therapy for the management of symptoms and quality of life in chronic pain patients.

Introduction

Chronic pain is one of the leading causes of disability worldwide, estimated to affect 15–30% of the adult population [1–3]. It poses considerable burden on the quality of life and psychosocial function of patients, as well as significant socioeconomic impacts [4, 5]. In the United States alone, the combined costs associated with health care and lost productivity have been estimated to exceed $500 billion per year [6].

For many chronic pain patients, conventional pharmaceutical medications are ineffective and/or not well tolerated. Opioid medications, in particular, have high abuse potential, questionable long-term efficacy, and have been associated with alarming rates of overdose and death [7–10]. In the United States, it has been estimated that 3.8 million adults misuse prescription opioid medications (POMs), which, due to their high abuse potential, can progress to heroin use [7, 10]. In 2015, POMs were associated with 15,000 deaths; in 2016, that figure more than doubled, with opioid misuse accounting for more than 40,000 deaths [10, 11].

In the midst of an ongoing opioid crisis, there is an urgent need for effective and safer treatment alternatives for chronic pain. With a high margin of safety and postulated regulatory effects of tetrahydrocannabinol (THC) and cannabidiol (CBD) on major endogenous pain circuitry systems, cannabis is emerging as a promising alternative or adjunctive treatment option for the management of chronic pain [12–15].

The analgesic effects of cannabis and cannabinoids have been demonstrated in preclinical as well as clinical studies, although the evidence has been at times inconclusive due to the heterogeneity of study designs, study populations, and study medications. Moreover, many clinical studies have been limited by relatively small samples and short duration and have mostly focused on neuropathic pain [15]. As such, there is a need for large-scale longitudinal studies to evaluate the effects of medical cannabis in chronic pain.

The aim of this study was to evaluate the short-term and long-term effects of plant-based medical cannabis (MC) on outcomes of interest related to pain, quality of life, tolerability, and opioid medication use in a large cohort of chronic pain patients using medical cannabis over the course of one year.

Methods

Study Design

This was a prospective, observational, longitudinal, open-label study design to evaluate the effectiveness of MC in the management of chronic pain symptomatology. Patients were recruited at Apollo Cannabis Clinics in Toronto, Canada, from October 2015 to March 2019. Patients who provided signed informed consent were asked to complete study questionnaires at baseline (before commencing treatment with MC) and then once a month for a total of 12 months (i.e., month 1 follow-up questionnaires, month 2 follow-up questionnaires, etc.). The study was approved by the Advarra Institutional Review Board and was conducted in accordance with research regulations, applicable international standards of Good Clinical Practice, institutional research policies and procedures, and the ethical principles laid out in the Declaration of Helsinki.

Study Population

The study population was comprised of chronic pain patients who were seeking treatment at Apollo Cannabis Clinics and were prescribed MC following an initial clinical evaluation of medical history and health status. Patients were included in the study based on the following inclusion criteria: 1) ≥25 years of age, 2) willing and able to provide informed consent and complete study assessments, 3) chronic pain for three or more months, and 4) newly prescribed MC for the treatment of chronic pain. Patients were excluded if they had used MC in the past three months.

Treatment Regimen

As part of the routine treatment process in the clinic, all patients underwent a clinical evaluation of medical history, health status, and symptomatology by the attending physician and were then advised on a cannabis-based treatment plan, as well as alternative non-cannabinoid-based treatment plans. The prescribing methodology included a personalized treatment plan with specific dosing instructions, route of administration, and a set THC limit, depending on individual medical histories, needs, and tolerability.

Outcomes of Interest

The baseline questionnaire captured demographic information such as age, sex, and medical history, the pain condition for which MC had been prescribed, and the duration of the condition. In addition, the following outcomes of interest were collected at baseline and follow-up:

Brief Pain Inventory

A self-administered rating scale that captures Pain Severity (0=“no pain” to 10=“pain as bad as you can imagine”) and Pain Interference (0=“does not interfere” to 10 =“completely interferes”) [16]. It has high psychometric value, with a Cronbach’s alpha of 0.84–0.94 [17], an intraclass correlation coefficient of 0.84–0.90, and a kappa value of <0.70 [18].

Short Form Health Survey

The Short Form Health Survey is a self-reported, multi-item scale questionnaire that evaluates quality of life. Derived from the SF-36, the 12-item Short Form Survey (SF-12) is a shorter version from which the Physical Composite Summary (PCS) and Mental Composite Summary (MCS) scores are calculated, ranging from 0=“lowest level of health” to 100=“highest level of health” [19, 20]. It has been reported to have a Cronbach’s alpha coefficient of 0.72–0.89 and a test–retest reliability of 0.73–0.86 [21].

Safety Measures

Study participants were asked to rate the frequency (“never,” “rarely,” “sometimes,” “frequently”) at which they experienced the following undesired symptoms at baseline and follow-up: fast heartbeat, dizziness, dry mouth, disorientation, increased appetite, drowsiness, feeling faint, fatigue, headache, impaired memory, confusion, feeling “high,” suspiciousness, nervousness, feelings of anxiety, paranoia, imbalance, throat irritation, hallucinations, impairment in motor skills, coughing, nausea, and vomiting.

Opioid Medications

Patients taking opioid medications were asked to provide the name and daily dose of opioid medications they were taking. In order to allow for comparisons across patients taking different types of opioid medications, reported daily opioid doses were converted into Oral Morphine Equivalents (OMEs), as described by Nielsen et al. [22].

Time Point Definition

Given that this was an observational study with no monetary incentives for completion of questionnaires, fewer participants completed their questionnaires at each subsequent monthly follow-up. To reduce the use of variables with missing data, analyses were conducted only on key time points—namely, baseline, month 1, month 3, month 6, and month 12 follow-up data. It should also be noted that many study participants did not complete the monthly follow-up questionnaires in a timely manner. As such, some patients completed their month 1 follow-up questionnaire at month 1 post–treatment initiation, while others may have completed it at month 3 post–treatment initiation. In order to make these data comparable regardless of when the questionnaires were completed, collected data were categorized into specific time points based on how many days had passed since the completion of the baseline report. As such, follow-up time points were defined as follows:

  • Baseline (B): day 0, before initiating MC treatment

  • Month 1 (M1): 20 to 54 days passed since completion of baseline report

  • Month 3 (M3): 85 to 114 days passed since completion of baseline report

  • Month 6 (M6): 175 to 204 days passed since completion of baseline report

  • Month 12 (M12): 355 to 385 days passed since completion of baseline report

Statistical Analysis

All statistical analyses were conducted using IBM SPSS Statistics for Mac (version 25.0; IBM Corp, Armonk, NY, USA) and XLSTAT Basic for Mac (Addinsoft, Long Island City, NY, USA). For the purposes of this manuscript, only the data collected at B, M1, M3, M6, and M12 were included in the analyses. Normally distributed continuous variables are presented as means with SDs, and non–normally distributed continuous variables are presented as medians, except where noted. Categorical variables are presented as counts and/or as valid percentages of the total. Continuous variables were checked for outliers by inspection of boxplots and for normality using the Shapiro-Wilk test. Changes over time in normally distributed continuous variables were analyzed with one-way repeated-measures analyses of variance (ANOVAs) and, where applicable, Bonferroni post hoc tests; changes over time in non–normally distributed continuous variables were analyzed with the Skillings-Mack test, a nonparametric test that is equivalent to the Friedman test, for use when there are missing data [23].

Exploratory analyses were conducted to evaluate differences between patients who completed all five time point visits (i.e., B, M1, M3, M6, and M12) and patients who completed only four time point visits (B, M1, M3, M6), patients who completed three time point visits (B, M1, M3), and those who completed only two time point visits (B and M1). Categorical variables are presented as valid percentages of the total, and associations between categorical variables, such as gender distribution across time point groups, were analyzed with chi-square tests.

Measures of safety, which comprised categorical response options, were converted into dichotomous variables, where the answer options “never” and “rarely” were combined into the category “never to rarely,” and the answer options “sometimes” to “frequently” were combined into the category “sometimes to frequently.” These variables were tested for the first and second assumptions of sample size adequacy and analyzed with Cochran’s Q test. Where applicable, McNemar’s post hoc tests with Bonferroni corrections were conducted on B, M1, M6, and M12, with statistical significance set at P < 0.0083 for six pairwise comparisons [24].

Results

Study Demographics

During the study period, a total of 1,245 patients provided informed consent and completed questionnaires. However, 494 (40%) completed only the baseline report but no other follow-up reports and, thus, were excluded from the analyses. Only patients who completed the baseline reports and at least one of the follow-up reports (i.e., month 1 and/or month 3 and/or month 6 and/or month 12) were included in the study sample, which resulted in a final study population of 751 (60%) at baseline.

The age of the patient population ranged from 25 to 88 years and was normally distributed, with a mean age of 49.6 ± 14.3 years. The sex distribution was fairly balanced, with 57% female patients.

Study participants reported a heterogeneous array of pain conditions, with the five most commonly reported conditions being back pain (including those with sciatica/neuropathic pain), osteoarthritis, chronic headaches, fibromyalgia, and degenerative disc disease (Table 1). Other less commonly reported pain conditions included pelvic pain, diabetic neuropathy, spinal cord injury, peripheral neuropathy, myofascial pain, and endometriosis, among others. The duration of these pain conditions ranged from three months to >10 years. Moreover, 47% of patients reported having one pain condition, while the rest reported suffering from two or more pain conditions (Table 1).

Table 1

Characteristics of pain conditions afflicting the study population at baseline

Primary pain condition, No. (% of intake total)Back pain351 (46.7)
Osteoarthritis214 (28.5)
Chronic headaches160 (21.3)
Fibromyalgia132 (17.6)
Degenerative disc disease123 (16.4)
Sciatica84 (11.2)
Rheumatoid arthritis81 (10.8)
Herniated disc73 (9.7)
Musculoskeletal pain72 (9.6)
Spinal stenosis56 (7.5)
No. of concurrent pain conditions, No. (%)1353 (47)
2–5364 (48.5)
6–1031 (4.1)
>103 (0.4)
Duration of pain conditions, No. (%)<6 mo74 (9.9)
6–11 mo26 (3.5)
1–5 y206 (27.4)
5–10 y173 (23)
Over 10 y272 (36.2)
Primary pain condition, No. (% of intake total)Back pain351 (46.7)
Osteoarthritis214 (28.5)
Chronic headaches160 (21.3)
Fibromyalgia132 (17.6)
Degenerative disc disease123 (16.4)
Sciatica84 (11.2)
Rheumatoid arthritis81 (10.8)
Herniated disc73 (9.7)
Musculoskeletal pain72 (9.6)
Spinal stenosis56 (7.5)
No. of concurrent pain conditions, No. (%)1353 (47)
2–5364 (48.5)
6–1031 (4.1)
>103 (0.4)
Duration of pain conditions, No. (%)<6 mo74 (9.9)
6–11 mo26 (3.5)
1–5 y206 (27.4)
5–10 y173 (23)
Over 10 y272 (36.2)
Table 1

Characteristics of pain conditions afflicting the study population at baseline

Primary pain condition, No. (% of intake total)Back pain351 (46.7)
Osteoarthritis214 (28.5)
Chronic headaches160 (21.3)
Fibromyalgia132 (17.6)
Degenerative disc disease123 (16.4)
Sciatica84 (11.2)
Rheumatoid arthritis81 (10.8)
Herniated disc73 (9.7)
Musculoskeletal pain72 (9.6)
Spinal stenosis56 (7.5)
No. of concurrent pain conditions, No. (%)1353 (47)
2–5364 (48.5)
6–1031 (4.1)
>103 (0.4)
Duration of pain conditions, No. (%)<6 mo74 (9.9)
6–11 mo26 (3.5)
1–5 y206 (27.4)
5–10 y173 (23)
Over 10 y272 (36.2)
Primary pain condition, No. (% of intake total)Back pain351 (46.7)
Osteoarthritis214 (28.5)
Chronic headaches160 (21.3)
Fibromyalgia132 (17.6)
Degenerative disc disease123 (16.4)
Sciatica84 (11.2)
Rheumatoid arthritis81 (10.8)
Herniated disc73 (9.7)
Musculoskeletal pain72 (9.6)
Spinal stenosis56 (7.5)
No. of concurrent pain conditions, No. (%)1353 (47)
2–5364 (48.5)
6–1031 (4.1)
>103 (0.4)
Duration of pain conditions, No. (%)<6 mo74 (9.9)
6–11 mo26 (3.5)
1–5 y206 (27.4)
5–10 y173 (23)
Over 10 y272 (36.2)

Prior Experience and Medical Cannabis Use at Follow-up

At baseline—before initiating MC treatment—60% of the study sample consisted of patients who had no previous experience with cannabis or had only used when younger (Table 2). Patients were prescribed medical cannabis with dosing instructions, route of administration, and set THC limits based on patient-specific needs and tolerability. The reported mean daily doses (in grams of dry flower per day or oil equivalent) were normally distributed at M1, M3, and M6 (P > 0.4), but not at M12 (P = 0.004), and did not differ significantly across time points (F(3,15)=0.798, P = 0.514) (Table 2). Data on exact THC and CBD doses were not collected in the study. However, the MC consumed by the study population typically ranged from 7% to 29% THC and/or CBD.

Table 2

Cannabis use before and after commencing

Cannabis experience at baseline, No. (% of intake total)No experience264 (35.2)
Used when younger187 (24.9)
Light to moderate user134 (17.8)
Frequent user130 (17.3)
Not reported36 (4.8)
Daily doses of MC, mean ± SD, g/dMonth 11.33 ± 0.41
Month 31.25 ± 0.52
Month 61.33 ± 0.61
Month 121.50 ± 0.55
Cannabis experience at baseline, No. (% of intake total)No experience264 (35.2)
Used when younger187 (24.9)
Light to moderate user134 (17.8)
Frequent user130 (17.3)
Not reported36 (4.8)
Daily doses of MC, mean ± SD, g/dMonth 11.33 ± 0.41
Month 31.25 ± 0.52
Month 61.33 ± 0.61
Month 121.50 ± 0.55

MC = medical cannabis.

Table 2

Cannabis use before and after commencing

Cannabis experience at baseline, No. (% of intake total)No experience264 (35.2)
Used when younger187 (24.9)
Light to moderate user134 (17.8)
Frequent user130 (17.3)
Not reported36 (4.8)
Daily doses of MC, mean ± SD, g/dMonth 11.33 ± 0.41
Month 31.25 ± 0.52
Month 61.33 ± 0.61
Month 121.50 ± 0.55
Cannabis experience at baseline, No. (% of intake total)No experience264 (35.2)
Used when younger187 (24.9)
Light to moderate user134 (17.8)
Frequent user130 (17.3)
Not reported36 (4.8)
Daily doses of MC, mean ± SD, g/dMonth 11.33 ± 0.41
Month 31.25 ± 0.52
Month 61.33 ± 0.61
Month 121.50 ± 0.55

MC = medical cannabis.

Correlates of Pain

The Brief Pain Inventory (BPI) was used to calculate Pain Interference (BPI-I) and Pain Severity (BPI-S), and changes over time were evaluated with one-way repeated-measures ANOVAs. There were no outliers in BPI-I scores, and the data were normally distributed (P > 0.1). BPI-S scores were also normally distributed across time points (P > 0.2), but there was one outlier at M1 and one outlier at M3. Both of these outliers were within the upper inner fence (i.e., mild outliers), and upon examination of each outlier, it was determined that there were no data collection, entry, or calculation errors associated with these scores. As such, both BPI-S outliers were included in the analyses.

The assumption of sphericity was met for both BPI-I and BPI-S scores, as assessed by Mauchly’s test of sphericity (χ2(9)=14.34, P = 0.112, and χ2(9)=6.30, P = 0.711, respectively). Treatment with MC was found to be associated with significant changes in BPI-I scores (F(4, 84)=8.99, P < 0.0005, partial η=0.30) and in BPI-S scores (F(4, 84)=9.98, P < 0.0005, partial η=0.32). Specifically, significant score reductions were observed from B to M1, B to M3, B to M6, and B to M12 (Table 3).

Table 3

Measures of pain interference and pain severity as per the Brief Pain Inventory

BPI-Interference
BPI-Severity
Mean ± SD95% CI, P ValueMean ± SD95% CI, P Value
Baseline (N=706)6.23 ± 1.635.58 ± 1.53
Month 1 (N=584)4.55 ± 2.39*0.48 to 2.88, 0.0034.27 ± 1.90*0.47 to 2.14, 0.001
Month 3 (N=230)4.08 ± 2.97*0.34 to 3.96, 0.0133.89 ± 2.17*0.55 to 2.83, 0.001
Month 6 (N=105)4.21 ± 2.64*0.45 to 3.58, 0.0063.99 ± 2.18*0.30 to 2.87, 0.009
Month 12 (N=43)3.54 ± 2.84*0.92 to 4.46, 0.0013.49 ± 2.17*0.90 to 3.27, <0.001
BPI-Interference
BPI-Severity
Mean ± SD95% CI, P ValueMean ± SD95% CI, P Value
Baseline (N=706)6.23 ± 1.635.58 ± 1.53
Month 1 (N=584)4.55 ± 2.39*0.48 to 2.88, 0.0034.27 ± 1.90*0.47 to 2.14, 0.001
Month 3 (N=230)4.08 ± 2.97*0.34 to 3.96, 0.0133.89 ± 2.17*0.55 to 2.83, 0.001
Month 6 (N=105)4.21 ± 2.64*0.45 to 3.58, 0.0063.99 ± 2.18*0.30 to 2.87, 0.009
Month 12 (N=43)3.54 ± 2.84*0.92 to 4.46, 0.0013.49 ± 2.17*0.90 to 3.27, <0.001

Treatment with medical cannabis was found to be associated with significant changes in Brief Pain Inventory measures of pain interference (F(4, 84)=8.99, P < 0.0005, partial η2 =0.30) and pain severity (F(4, 84)=9.98, P < 0.0005, partial η2 =0.32).

BPI = Brief Pain Inventory; CI = confidence interval.

*

Denotes statistical significance in relation to baseline.

Table 3

Measures of pain interference and pain severity as per the Brief Pain Inventory

BPI-Interference
BPI-Severity
Mean ± SD95% CI, P ValueMean ± SD95% CI, P Value
Baseline (N=706)6.23 ± 1.635.58 ± 1.53
Month 1 (N=584)4.55 ± 2.39*0.48 to 2.88, 0.0034.27 ± 1.90*0.47 to 2.14, 0.001
Month 3 (N=230)4.08 ± 2.97*0.34 to 3.96, 0.0133.89 ± 2.17*0.55 to 2.83, 0.001
Month 6 (N=105)4.21 ± 2.64*0.45 to 3.58, 0.0063.99 ± 2.18*0.30 to 2.87, 0.009
Month 12 (N=43)3.54 ± 2.84*0.92 to 4.46, 0.0013.49 ± 2.17*0.90 to 3.27, <0.001
BPI-Interference
BPI-Severity
Mean ± SD95% CI, P ValueMean ± SD95% CI, P Value
Baseline (N=706)6.23 ± 1.635.58 ± 1.53
Month 1 (N=584)4.55 ± 2.39*0.48 to 2.88, 0.0034.27 ± 1.90*0.47 to 2.14, 0.001
Month 3 (N=230)4.08 ± 2.97*0.34 to 3.96, 0.0133.89 ± 2.17*0.55 to 2.83, 0.001
Month 6 (N=105)4.21 ± 2.64*0.45 to 3.58, 0.0063.99 ± 2.18*0.30 to 2.87, 0.009
Month 12 (N=43)3.54 ± 2.84*0.92 to 4.46, 0.0013.49 ± 2.17*0.90 to 3.27, <0.001

Treatment with medical cannabis was found to be associated with significant changes in Brief Pain Inventory measures of pain interference (F(4, 84)=8.99, P < 0.0005, partial η2 =0.30) and pain severity (F(4, 84)=9.98, P < 0.0005, partial η2 =0.32).

BPI = Brief Pain Inventory; CI = confidence interval.

*

Denotes statistical significance in relation to baseline.

The scores of the individual domains of the BPI-I subscale (i.e., General Activity, Mood, Walking Ability, Normal Work, Relationships with Other People, Sleep, and Enjoyment of Life) were also examined. Shapiro Wilk’s tests indicated that BPI-I subdomain scores were not normally distributed across all the time points, and thus they were analyzed with Skillings-Mack tests. All the subscores of the BPI-I domain were found to have a computed P value <0.01, indicating statistically significant improvements over the time. A detailed breakdown of the BPI-I subscores at each time point of interest is displayed in the Supplementary Data.

Moreover, given that a proportion of patients reported cannabis use before initiation of MC treatment, two-way repeated-measures ANOVAs were also conducted to evaluate the effects of time and prior cannabis experience/frequency of use on BPI scores. As expected based on the results presented in Table 3, there were statistically significant changes in BPI-I (F(4,72)=9.90, P < 0.0001) and BPI-S scores (F(4,72)=9.94, P < 0.0001) over the course of the treatment observation period. However, there were no statistically significant interactions between time and prior cannabis experience/frequency of use for either BPI-I (F(12,72)=0.79, P = 0.655) or BPI-S (F(12,72)=0.78, P = 0.668) (Supplementary Data).

Quality of Life

The PCS and MCS scores derived from the SF-12 were used as a measure of quality of life. Inspection of scores revealed one upper inner fence and one upper outer fence PCS outliers at M1, two upper inner fence PCS outliers at M3, and one lower inner fence MCS outlier at M6. Each outlier was inspected and determined not to be associated with data collection, entry, or calculation errors, and as such, all five outliers were included in the analyses. Shapiro Wilk’s tests indicated that PCS and MCS data were not normally distributed across all time points of interest. As such, score changes over time were evaluated with Skillings-Mack tests. For both PCS and MCS scores, the computed P values were <0.05, indicating statistically significant improvements over time (Table 4).

Table 4

Physical and mental health–related measures of quality of life as per the Short Form Health Survey

Mean ± SDMedianSum of RanksMean of RanksTest Statistics
Physical Composite Summary scoreBaseline (N=509)31.21 ± 8.0930.189112
  • Q (observed value) = 18

  • Q (critical value) = 9

  • DF = 4

  • P < 0.05

Month 1 (N=435)32.99 ± 9.9031.109922
Month 3 (N= 148)34.05 ± 9.4231.809937
Month 6 (N=69)34.44 ± 10.7931.7198614
Month 12 (N=31)33.12 ± 11.1231.2697732
Mental Composite Summary scoreBaseline (N=509)42.83 ± 11.5341.599152
  • Q (observed value) = 17

  • Q (critical value) = 9

  • DF = 4

  • P < 0.05

Month 1 (N=435)46.55 ± 11.3948.121,0062
Month 3 (N=148)47.26 ± 11.2348.409897
Month 6 (N=69)45.36 ± 11.7445.9297314
Month 12 (N=31)51.05 ± 9.4250.0997631
Mean ± SDMedianSum of RanksMean of RanksTest Statistics
Physical Composite Summary scoreBaseline (N=509)31.21 ± 8.0930.189112
  • Q (observed value) = 18

  • Q (critical value) = 9

  • DF = 4

  • P < 0.05

Month 1 (N=435)32.99 ± 9.9031.109922
Month 3 (N= 148)34.05 ± 9.4231.809937
Month 6 (N=69)34.44 ± 10.7931.7198614
Month 12 (N=31)33.12 ± 11.1231.2697732
Mental Composite Summary scoreBaseline (N=509)42.83 ± 11.5341.599152
  • Q (observed value) = 17

  • Q (critical value) = 9

  • DF = 4

  • P < 0.05

Month 1 (N=435)46.55 ± 11.3948.121,0062
Month 3 (N=148)47.26 ± 11.2348.409897
Month 6 (N=69)45.36 ± 11.7445.9297314
Month 12 (N=31)51.05 ± 9.4250.0997631

Treatment with medical cannabis was found to be associated with significant changes in the Physical and Mental Health domains of the SF-12 over the course of the 12-month observation period.

DF = degrees of freedom; MCS = Mental Composite Summary; PCS = Physical Composite Summary; SF-12 = Short Form Health Survey.

Table 4

Physical and mental health–related measures of quality of life as per the Short Form Health Survey

Mean ± SDMedianSum of RanksMean of RanksTest Statistics
Physical Composite Summary scoreBaseline (N=509)31.21 ± 8.0930.189112
  • Q (observed value) = 18

  • Q (critical value) = 9

  • DF = 4

  • P < 0.05

Month 1 (N=435)32.99 ± 9.9031.109922
Month 3 (N= 148)34.05 ± 9.4231.809937
Month 6 (N=69)34.44 ± 10.7931.7198614
Month 12 (N=31)33.12 ± 11.1231.2697732
Mental Composite Summary scoreBaseline (N=509)42.83 ± 11.5341.599152
  • Q (observed value) = 17

  • Q (critical value) = 9

  • DF = 4

  • P < 0.05

Month 1 (N=435)46.55 ± 11.3948.121,0062
Month 3 (N=148)47.26 ± 11.2348.409897
Month 6 (N=69)45.36 ± 11.7445.9297314
Month 12 (N=31)51.05 ± 9.4250.0997631
Mean ± SDMedianSum of RanksMean of RanksTest Statistics
Physical Composite Summary scoreBaseline (N=509)31.21 ± 8.0930.189112
  • Q (observed value) = 18

  • Q (critical value) = 9

  • DF = 4

  • P < 0.05

Month 1 (N=435)32.99 ± 9.9031.109922
Month 3 (N= 148)34.05 ± 9.4231.809937
Month 6 (N=69)34.44 ± 10.7931.7198614
Month 12 (N=31)33.12 ± 11.1231.2697732
Mental Composite Summary scoreBaseline (N=509)42.83 ± 11.5341.599152
  • Q (observed value) = 17

  • Q (critical value) = 9

  • DF = 4

  • P < 0.05

Month 1 (N=435)46.55 ± 11.3948.121,0062
Month 3 (N=148)47.26 ± 11.2348.409897
Month 6 (N=69)45.36 ± 11.7445.9297314
Month 12 (N=31)51.05 ± 9.4250.0997631

Treatment with medical cannabis was found to be associated with significant changes in the Physical and Mental Health domains of the SF-12 over the course of the 12-month observation period.

DF = degrees of freedom; MCS = Mental Composite Summary; PCS = Physical Composite Summary; SF-12 = Short Form Health Survey.

Adverse Events

Study patients were asked to rate the frequency at which they experienced undesired symptoms commonly associated with cannabis use (Table 5, Figure 1).

Frequencies of select adverse effects at baseline and over the course of the study observation period. A) There were no statistically significant changes in the frequencies at which study subjects experienced adverse events commonly associated with cannabis use, such as dry mouth, increased appetite, drowsiness, or impaired memory. B) There were statistically significant reductions in the frequencies at which study subjects experienced fatigue, headaches, feelings of anxiety, and nausea. A full breakdown of adverse event frequencies is displayed in Table 5. *Statistical significance in relation to baseline.
Figure 1

Frequencies of select adverse effects at baseline and over the course of the study observation period. A) There were no statistically significant changes in the frequencies at which study subjects experienced adverse events commonly associated with cannabis use, such as dry mouth, increased appetite, drowsiness, or impaired memory. B) There were statistically significant reductions in the frequencies at which study subjects experienced fatigue, headaches, feelings of anxiety, and nausea. A full breakdown of adverse event frequencies is displayed in Table 5. *Statistical significance in relation to baseline.

Table 5

Frequency of adverse events at baseline and over the course of the 12-month study observation period

Baseline (N = 751), %Month 1 (N = 628), %Month 3 (N = 248), %Month 6 (N = 112), %Month 12 (N = 46), %
Fast heartbeatNever to rarely75.280.983.585.789.1
Sometimes to frequently24.819.116.514.310.9
DizzinessNever to rarely69.173.974.282.182.6
Sometimes to frequently30.926.125.817.917.4
Dry mouthNever to rarely53.445.247.245.545.7
Sometimes to frequently46.654.852.854.554.3
DisorientationNever to rarely88.890.491.596.493.5
Sometimes to frequently11.29.68.53.66.5
Increased appetiteNever to rarely64.354.554.460.743.5
Sometimes to frequently35.745.545.639.356.5
DrowsinessNever to rarely50.150.64850.965.2
Sometimes to frequently49.949.45249.134.8
Feeling faintNever to rarely84.289.689.593.895.7
Sometimes to frequently15.810.410.56.34.3
FatigueNever to rarely2443.344.85052.2
Sometimes to frequently7656.7*55.350*47.8*
HeadacheNever to rarely40.760.456.562.558.7
Sometimes to frequently59.339.6*43.537.541.3
Impaired memoryNever to rarely6576.476.277.782.6
Sometimes to frequently3523.623.822.317.4
ConfusionNever to rarely829090.392.991.3
Sometimes to frequently18109.77.18.7
Feeling highNever to rarely82.869.466.968.865.2
Sometimes to frequently17.230.633.131.334.8
SuspiciousnessNever to rarely91.695.297.698.297.8
Sometimes to frequently8.44.82.41.82.2
NervousnessNever to rarely6682.580.683.993.5
Sometimes to frequently3417.419.416.16.5
Feelings of anxietyNever to rarely62.378.78183.993.5
Sometimes to frequently37.721.3*1916.1*6.5*
ParanoiaNever to rarely94.896.596.897.3100
Sometimes to frequently5.23.53.22.70
ImbalanceNever to rarely75.884.287.585.784.8
Sometimes to frequently24.215.812.514.315.2
Throat irritationNever to rarely83.88380.672.376.1
Sometimes to frequently16.21719.427.723.9
HallucinationsNever to rarely98.899.29898.297.8
Sometimes to frequently1.20.821.82.2
Impaired motor skillsNever to rarely8792.792.387.591.3
Sometimes to frequently137.37.712.58.7
CoughingNever to rarely71.475.8797576.1
Sometimes to frequently28.624.2212523.9
NauseaNever to rarely71.283.385.182.189.1
Sometimes to frequently28.816.7*14.917.9*10.9
VomitingNever to rarely92.59796.895.595.7
Sometimes to frequently7.533.24.54.3
Baseline (N = 751), %Month 1 (N = 628), %Month 3 (N = 248), %Month 6 (N = 112), %Month 12 (N = 46), %
Fast heartbeatNever to rarely75.280.983.585.789.1
Sometimes to frequently24.819.116.514.310.9
DizzinessNever to rarely69.173.974.282.182.6
Sometimes to frequently30.926.125.817.917.4
Dry mouthNever to rarely53.445.247.245.545.7
Sometimes to frequently46.654.852.854.554.3
DisorientationNever to rarely88.890.491.596.493.5
Sometimes to frequently11.29.68.53.66.5
Increased appetiteNever to rarely64.354.554.460.743.5
Sometimes to frequently35.745.545.639.356.5
DrowsinessNever to rarely50.150.64850.965.2
Sometimes to frequently49.949.45249.134.8
Feeling faintNever to rarely84.289.689.593.895.7
Sometimes to frequently15.810.410.56.34.3
FatigueNever to rarely2443.344.85052.2
Sometimes to frequently7656.7*55.350*47.8*
HeadacheNever to rarely40.760.456.562.558.7
Sometimes to frequently59.339.6*43.537.541.3
Impaired memoryNever to rarely6576.476.277.782.6
Sometimes to frequently3523.623.822.317.4
ConfusionNever to rarely829090.392.991.3
Sometimes to frequently18109.77.18.7
Feeling highNever to rarely82.869.466.968.865.2
Sometimes to frequently17.230.633.131.334.8
SuspiciousnessNever to rarely91.695.297.698.297.8
Sometimes to frequently8.44.82.41.82.2
NervousnessNever to rarely6682.580.683.993.5
Sometimes to frequently3417.419.416.16.5
Feelings of anxietyNever to rarely62.378.78183.993.5
Sometimes to frequently37.721.3*1916.1*6.5*
ParanoiaNever to rarely94.896.596.897.3100
Sometimes to frequently5.23.53.22.70
ImbalanceNever to rarely75.884.287.585.784.8
Sometimes to frequently24.215.812.514.315.2
Throat irritationNever to rarely83.88380.672.376.1
Sometimes to frequently16.21719.427.723.9
HallucinationsNever to rarely98.899.29898.297.8
Sometimes to frequently1.20.821.82.2
Impaired motor skillsNever to rarely8792.792.387.591.3
Sometimes to frequently137.37.712.58.7
CoughingNever to rarely71.475.8797576.1
Sometimes to frequently28.624.2212523.9
NauseaNever to rarely71.283.385.182.189.1
Sometimes to frequently28.816.7*14.917.9*10.9
VomitingNever to rarely92.59796.895.595.7
Sometimes to frequently7.533.24.54.3

There were statistically significant reductions in the frequencies of fatigue (Q = 16.12, P = 0.002), headaches (Q = 10.51, P = 0.034), feelings of anxiety (Q = 15.85, P = 0.003), and nausea (Q = 17.04, P = 0.001). Bolded lines refer to AEs that demonstrated significant changes over time.

*

Denotes statistical significance in relation to baseline.

Table 5

Frequency of adverse events at baseline and over the course of the 12-month study observation period

Baseline (N = 751), %Month 1 (N = 628), %Month 3 (N = 248), %Month 6 (N = 112), %Month 12 (N = 46), %
Fast heartbeatNever to rarely75.280.983.585.789.1
Sometimes to frequently24.819.116.514.310.9
DizzinessNever to rarely69.173.974.282.182.6
Sometimes to frequently30.926.125.817.917.4
Dry mouthNever to rarely53.445.247.245.545.7
Sometimes to frequently46.654.852.854.554.3
DisorientationNever to rarely88.890.491.596.493.5
Sometimes to frequently11.29.68.53.66.5
Increased appetiteNever to rarely64.354.554.460.743.5
Sometimes to frequently35.745.545.639.356.5
DrowsinessNever to rarely50.150.64850.965.2
Sometimes to frequently49.949.45249.134.8
Feeling faintNever to rarely84.289.689.593.895.7
Sometimes to frequently15.810.410.56.34.3
FatigueNever to rarely2443.344.85052.2
Sometimes to frequently7656.7*55.350*47.8*
HeadacheNever to rarely40.760.456.562.558.7
Sometimes to frequently59.339.6*43.537.541.3
Impaired memoryNever to rarely6576.476.277.782.6
Sometimes to frequently3523.623.822.317.4
ConfusionNever to rarely829090.392.991.3
Sometimes to frequently18109.77.18.7
Feeling highNever to rarely82.869.466.968.865.2
Sometimes to frequently17.230.633.131.334.8
SuspiciousnessNever to rarely91.695.297.698.297.8
Sometimes to frequently8.44.82.41.82.2
NervousnessNever to rarely6682.580.683.993.5
Sometimes to frequently3417.419.416.16.5
Feelings of anxietyNever to rarely62.378.78183.993.5
Sometimes to frequently37.721.3*1916.1*6.5*
ParanoiaNever to rarely94.896.596.897.3100
Sometimes to frequently5.23.53.22.70
ImbalanceNever to rarely75.884.287.585.784.8
Sometimes to frequently24.215.812.514.315.2
Throat irritationNever to rarely83.88380.672.376.1
Sometimes to frequently16.21719.427.723.9
HallucinationsNever to rarely98.899.29898.297.8
Sometimes to frequently1.20.821.82.2
Impaired motor skillsNever to rarely8792.792.387.591.3
Sometimes to frequently137.37.712.58.7
CoughingNever to rarely71.475.8797576.1
Sometimes to frequently28.624.2212523.9
NauseaNever to rarely71.283.385.182.189.1
Sometimes to frequently28.816.7*14.917.9*10.9
VomitingNever to rarely92.59796.895.595.7
Sometimes to frequently7.533.24.54.3
Baseline (N = 751), %Month 1 (N = 628), %Month 3 (N = 248), %Month 6 (N = 112), %Month 12 (N = 46), %
Fast heartbeatNever to rarely75.280.983.585.789.1
Sometimes to frequently24.819.116.514.310.9
DizzinessNever to rarely69.173.974.282.182.6
Sometimes to frequently30.926.125.817.917.4
Dry mouthNever to rarely53.445.247.245.545.7
Sometimes to frequently46.654.852.854.554.3
DisorientationNever to rarely88.890.491.596.493.5
Sometimes to frequently11.29.68.53.66.5
Increased appetiteNever to rarely64.354.554.460.743.5
Sometimes to frequently35.745.545.639.356.5
DrowsinessNever to rarely50.150.64850.965.2
Sometimes to frequently49.949.45249.134.8
Feeling faintNever to rarely84.289.689.593.895.7
Sometimes to frequently15.810.410.56.34.3
FatigueNever to rarely2443.344.85052.2
Sometimes to frequently7656.7*55.350*47.8*
HeadacheNever to rarely40.760.456.562.558.7
Sometimes to frequently59.339.6*43.537.541.3
Impaired memoryNever to rarely6576.476.277.782.6
Sometimes to frequently3523.623.822.317.4
ConfusionNever to rarely829090.392.991.3
Sometimes to frequently18109.77.18.7
Feeling highNever to rarely82.869.466.968.865.2
Sometimes to frequently17.230.633.131.334.8
SuspiciousnessNever to rarely91.695.297.698.297.8
Sometimes to frequently8.44.82.41.82.2
NervousnessNever to rarely6682.580.683.993.5
Sometimes to frequently3417.419.416.16.5
Feelings of anxietyNever to rarely62.378.78183.993.5
Sometimes to frequently37.721.3*1916.1*6.5*
ParanoiaNever to rarely94.896.596.897.3100
Sometimes to frequently5.23.53.22.70
ImbalanceNever to rarely75.884.287.585.784.8
Sometimes to frequently24.215.812.514.315.2
Throat irritationNever to rarely83.88380.672.376.1
Sometimes to frequently16.21719.427.723.9
HallucinationsNever to rarely98.899.29898.297.8
Sometimes to frequently1.20.821.82.2
Impaired motor skillsNever to rarely8792.792.387.591.3
Sometimes to frequently137.37.712.58.7
CoughingNever to rarely71.475.8797576.1
Sometimes to frequently28.624.2212523.9
NauseaNever to rarely71.283.385.182.189.1
Sometimes to frequently28.816.7*14.917.9*10.9
VomitingNever to rarely92.59796.895.595.7
Sometimes to frequently7.533.24.54.3

There were statistically significant reductions in the frequencies of fatigue (Q = 16.12, P = 0.002), headaches (Q = 10.51, P = 0.034), feelings of anxiety (Q = 15.85, P = 0.003), and nausea (Q = 17.04, P = 0.001). Bolded lines refer to AEs that demonstrated significant changes over time.

*

Denotes statistical significance in relation to baseline.

Table 6

Opioid medication doses, measures of pain, and quality of life in a subset of patients using opioid medications at baseline

Mean ± SDSum of RanksMean of RanksTest Statistics
OME*Baseline (N =82)26.23 ± 48.131642
  • Q (observed) = 26

  • Q (critical) = 9

  • DF = 4

  • P<0.0001

Month 1 (N =67)12.69 ± 44.701192
Month 3 (N =26)3.28 ± 8.621285
Month 6 (N =9)3.00 ± 6.5013115
Month 12 (N =4)1.38 ± 0.9713434
BPI-S*Baseline (N =76)5.77 ± 1.771502
  • Q (observed) = 15

  • Q (critical) = 9

  • DF = 4

  • P = 0.01

Month 1 (N =65)4.64 ± 2.231172
Month 3 (N =23)4.58 ± 2.111234
Month 6 (N =9)5.19 ± 2.2113014
Month 12 (N =4)4.06 ± 3.1612832
BPI-I*Baseline (N =76)6.40 ± 2.391522
  • Q (observed) = 21

  • Q (critical) = 9

  • DF = 4

  • P = 0.0003

Month 1 (N =65)5.00 ± 2.871242
Month 3 (N =23)4.41 ± 2.811155
Month 6 (N =9)5.36 ± 2.9513115
Month 12 (N =4)3.86 ± 3.2612926
SF-12 PCSBaseline (N =61)30.24 ± 8.851042
  • Q (observed) = 5

  • Q (critical) = 9

  • DF = 4

  • P = 0.29

Month 1 (N =49)31.54 ± 11.221062
Month 3 (N =17)34.31 ± 8.771147
Month 6 (N =7)34.18 ± 10.3711316
Month 12 (N =3)32.40 ± 8.6511237
SF-12 MCSBaseline (N =61)42.10 ± 11.461062
  • Q (observed) = 3

  • Q (critical) = 9

  • DF = 4

  • P=0.48

Month 1 (N =49)47.21 ± 12.751162
Month 3 (N =17)46.14 ± 13.841127
Month 6 (N =7)41.62 ± 12.7610515
Month 12 (N =3)52.68 ± 7.8511037
Mean ± SDSum of RanksMean of RanksTest Statistics
OME*Baseline (N =82)26.23 ± 48.131642
  • Q (observed) = 26

  • Q (critical) = 9

  • DF = 4

  • P<0.0001

Month 1 (N =67)12.69 ± 44.701192
Month 3 (N =26)3.28 ± 8.621285
Month 6 (N =9)3.00 ± 6.5013115
Month 12 (N =4)1.38 ± 0.9713434
BPI-S*Baseline (N =76)5.77 ± 1.771502
  • Q (observed) = 15

  • Q (critical) = 9

  • DF = 4

  • P = 0.01

Month 1 (N =65)4.64 ± 2.231172
Month 3 (N =23)4.58 ± 2.111234
Month 6 (N =9)5.19 ± 2.2113014
Month 12 (N =4)4.06 ± 3.1612832
BPI-I*Baseline (N =76)6.40 ± 2.391522
  • Q (observed) = 21

  • Q (critical) = 9

  • DF = 4

  • P = 0.0003

Month 1 (N =65)5.00 ± 2.871242
Month 3 (N =23)4.41 ± 2.811155
Month 6 (N =9)5.36 ± 2.9513115
Month 12 (N =4)3.86 ± 3.2612926
SF-12 PCSBaseline (N =61)30.24 ± 8.851042
  • Q (observed) = 5

  • Q (critical) = 9

  • DF = 4

  • P = 0.29

Month 1 (N =49)31.54 ± 11.221062
Month 3 (N =17)34.31 ± 8.771147
Month 6 (N =7)34.18 ± 10.3711316
Month 12 (N =3)32.40 ± 8.6511237
SF-12 MCSBaseline (N =61)42.10 ± 11.461062
  • Q (observed) = 3

  • Q (critical) = 9

  • DF = 4

  • P=0.48

Month 1 (N =49)47.21 ± 12.751162
Month 3 (N =17)46.14 ± 13.841127
Month 6 (N =7)41.62 ± 12.7610515
Month 12 (N =3)52.68 ± 7.8511037

Opioid medication doses were converted to oral morphine equivalents in mg/d to allow for comparisons across different opioid medication types.

BPI-I = Brief Pain Inventory–Pain Interference; BPI-S = Brief Pain Inventory–Pain Severity; SF-12 PCS = Short Form Health Survey–Physical Composite Summary Score; SF-12 MCS = Short Form Health Survey–Mental Composite Summary Score.

Table 6

Opioid medication doses, measures of pain, and quality of life in a subset of patients using opioid medications at baseline

Mean ± SDSum of RanksMean of RanksTest Statistics
OME*Baseline (N =82)26.23 ± 48.131642
  • Q (observed) = 26

  • Q (critical) = 9

  • DF = 4

  • P<0.0001

Month 1 (N =67)12.69 ± 44.701192
Month 3 (N =26)3.28 ± 8.621285
Month 6 (N =9)3.00 ± 6.5013115
Month 12 (N =4)1.38 ± 0.9713434
BPI-S*Baseline (N =76)5.77 ± 1.771502
  • Q (observed) = 15

  • Q (critical) = 9

  • DF = 4

  • P = 0.01

Month 1 (N =65)4.64 ± 2.231172
Month 3 (N =23)4.58 ± 2.111234
Month 6 (N =9)5.19 ± 2.2113014
Month 12 (N =4)4.06 ± 3.1612832
BPI-I*Baseline (N =76)6.40 ± 2.391522
  • Q (observed) = 21

  • Q (critical) = 9

  • DF = 4

  • P = 0.0003

Month 1 (N =65)5.00 ± 2.871242
Month 3 (N =23)4.41 ± 2.811155
Month 6 (N =9)5.36 ± 2.9513115
Month 12 (N =4)3.86 ± 3.2612926
SF-12 PCSBaseline (N =61)30.24 ± 8.851042
  • Q (observed) = 5

  • Q (critical) = 9

  • DF = 4

  • P = 0.29

Month 1 (N =49)31.54 ± 11.221062
Month 3 (N =17)34.31 ± 8.771147
Month 6 (N =7)34.18 ± 10.3711316
Month 12 (N =3)32.40 ± 8.6511237
SF-12 MCSBaseline (N =61)42.10 ± 11.461062
  • Q (observed) = 3

  • Q (critical) = 9

  • DF = 4

  • P=0.48

Month 1 (N =49)47.21 ± 12.751162
Month 3 (N =17)46.14 ± 13.841127
Month 6 (N =7)41.62 ± 12.7610515
Month 12 (N =3)52.68 ± 7.8511037
Mean ± SDSum of RanksMean of RanksTest Statistics
OME*Baseline (N =82)26.23 ± 48.131642
  • Q (observed) = 26

  • Q (critical) = 9

  • DF = 4

  • P<0.0001

Month 1 (N =67)12.69 ± 44.701192
Month 3 (N =26)3.28 ± 8.621285
Month 6 (N =9)3.00 ± 6.5013115
Month 12 (N =4)1.38 ± 0.9713434
BPI-S*Baseline (N =76)5.77 ± 1.771502
  • Q (observed) = 15

  • Q (critical) = 9

  • DF = 4

  • P = 0.01

Month 1 (N =65)4.64 ± 2.231172
Month 3 (N =23)4.58 ± 2.111234
Month 6 (N =9)5.19 ± 2.2113014
Month 12 (N =4)4.06 ± 3.1612832
BPI-I*Baseline (N =76)6.40 ± 2.391522
  • Q (observed) = 21

  • Q (critical) = 9

  • DF = 4

  • P = 0.0003

Month 1 (N =65)5.00 ± 2.871242
Month 3 (N =23)4.41 ± 2.811155
Month 6 (N =9)5.36 ± 2.9513115
Month 12 (N =4)3.86 ± 3.2612926
SF-12 PCSBaseline (N =61)30.24 ± 8.851042
  • Q (observed) = 5

  • Q (critical) = 9

  • DF = 4

  • P = 0.29

Month 1 (N =49)31.54 ± 11.221062
Month 3 (N =17)34.31 ± 8.771147
Month 6 (N =7)34.18 ± 10.3711316
Month 12 (N =3)32.40 ± 8.6511237
SF-12 MCSBaseline (N =61)42.10 ± 11.461062
  • Q (observed) = 3

  • Q (critical) = 9

  • DF = 4

  • P=0.48

Month 1 (N =49)47.21 ± 12.751162
Month 3 (N =17)46.14 ± 13.841127
Month 6 (N =7)41.62 ± 12.7610515
Month 12 (N =3)52.68 ± 7.8511037

Opioid medication doses were converted to oral morphine equivalents in mg/d to allow for comparisons across different opioid medication types.

BPI-I = Brief Pain Inventory–Pain Interference; BPI-S = Brief Pain Inventory–Pain Severity; SF-12 PCS = Short Form Health Survey–Physical Composite Summary Score; SF-12 MCS = Short Form Health Survey–Mental Composite Summary Score.

At M1, which was the first data collection time point after initiating MC treatment, the most commonly reported adverse events (e.g., reported as “sometimes to frequently” in at least 40% of the study sample) were dry mouth (reported by 54.8% of the study sample), increased appetite (reported by 45.5%), and drowsiness (reported by 49.4%). However, the frequencies at which these symptoms were reported at M1 were not statistically different from baseline.

On the other hand, there were statistically significant changes in the frequencies at which patients experienced fatigue (Q = 16.12, P = 0.002), headaches (Q = 10.51, P = 0.034), feelings of anxiety (Q = 15.85, P = 0.003), and nausea (Q = 17.04, P = 0.001). Post hoc analyses revealed that, compared with baseline, the frequencies of these symptoms decreased at follow-up. Specifically, the proportions of patients reporting fatigue decreased from B to M1, B to M6, and B to M12 (all at P ≤ 0.002); headaches decreased from B to M1 (P < 0.0005); feelings of anxiety decreased from B to M1, B to M6, and B to M12 (all at P < 0.0005); and nausea decreased from B to M1 and B to M6 (both at P ≤ 0.007).

Exploratory Analyses

Opioid Medication Use

Of the 751 study participants included in the analysis, a subset of patients reported taking opioid medications at baseline. This subset consisted of 82 patients, with an age range of 26 to 76 years and a mean age of 50.7 ± 13.4 years. Similar to the full study population, gender distribution was fairly balanced, with 63% females (N = 51). As noted in Time Point Definition, due to the observational nature of this study, each subsequent time point had fewer report completions. For this subset of patients, there were 82 report completions at baseline, 67 report completions at M1, 26 at M3, nine at M6, and four at M12.

Reported daily doses of opioid medications were converted to OMEs as described in Nielsen et al. (2016). Descriptions and tests statistics are displayed in Table 6. These data were not normally distributed and were thus analyzed with the Skillings-Mack test [23], which revealed statistically significant reductions on OME doses over the course of the treatment observation period.

Measures of pain and quality of life were also analyzed with the Skillings-Mack test due to non-normal distribution. These analyses revealed significant improvements in pain severity and pain interference, but not on the SF-12 measures of physical and mental health (Table 6).

Time Point Group Comparisons

Given the high rate of patients lost to follow-up at each subsequent visit, we sought to explore potential differences between patients who completed all five time point visits (i.e., B, M1, M3, M6, and M12; hereinafter referred to as the 5-TMP group) and patients who completed four time points (i.e., B, M1, M3, and M6; or 4-TMP), three time points (i.e., B, M1, and M3; or 3-TMP), or two time points (B and M1 only; or 2-TMP). There were no significant differences in baseline measures between patients who completed all five time point visits and patients who did not (Supplementary Data).

In addition, we evaluated the pattern of treatment response in patients in the 5-TMP, 4-TMP, 3-TMP, and 2-TMP groups by conducting individual repeated-measures ANOVAs on pain scores in each of these patient groups. These analyses were specifically conducted because repeated-measures ANOVAs are tests that exclude cases listwise; that is, only subjects who complete all the time points of interest are included in the analysis. For instance, a patient who completed only the B, M6, and M12 reports would be automatically excluded from an analysis in the 5-TMP group, because M1 and M3 reports were not completed. The BPI results presented in Correlates of Pain are based on analyses of patients who had completed all five time point reports, which included 22 patients only. As this is a small sample size, which considerably limits the generalizability of results, we sought to examine whether the pattern of response observed in the 5-TMP group was comparable to the pattern of response observed in the 4-TMP group (which included 64 patients), 3-TMP group (N = 184), and the 2-TMP group (N = 580). These analyses revealed similar patterns of response across time point groups, up to the last time point included in the analysis. That is, the 5-TMP group had significant changes in BPI scores from B to M1, B to M3, B to M6, and B to M12; the 4-TMP group had significant changes from B to M1, B to M3, and B to M6; the 3-TMP group had significant changes from B to M1 and B to M3; and the 2-TMP group had significant changes from B to M1 (Supplementary Data).

Discussion

Pain Management and Quality of Life

In this prospective, longitudinal, observational study of chronic pain patients, treatment with medical cannabis was associated with significant improvements in measures of pain and health-related quality of life.

Significant improvements in pain severity and pain interference were observed as early as one month after initiating treatment and were maintained over the course of the 12-month observation period during which patients who remained in the study were followed. The mean BPI-S score was 5.58 ± 1.53 at baseline, indicating moderate to severe pain, and 3.49 ± 2.17 at M12, indicating mild to moderate pain [18, 25, 26]. The 2.09-point reduction in pain severity over the course of the treatment observation period is indicative of a clinically significant improvement [27].

Although the analgesic effects of MC have been demonstrated in short-term studies of neuropathic pain [28–32], as well as in capsaicin-induced pain and hyperalgesia in healthy volunteers [33], fewer studies have been published on the long-term effects, particularly in non-neuropathic chronic pain. A PubMed search conducted at the time of the study, as well as inspection of reference lists in review articles and meta-analyses, revealed four other published long-term studies on the effects of plant-based cannabis in chronic pain patients. Bellneier et al. [34] conducted a three-month retrospective study of patients using THC/CBD oil capsules. Haroutounian et al. [35] conducted a six-month prospective open-label study of 206 patients using individually optimized doses of MC, and Ware et al. [36] conducted a one-year prospective cohort study of 215 patients using optimized doses of MC. In line with our findings, these studies found significant improvements in correlates of pain. On the other hand, Campbell et al. [15] conducted a large (N ≥ 1,000) four-year prospective cohort study and found no changes in measures of pain associated with cannabis. It should be noted, however, that participants in the study by Campbell et al. [15] had obtained illicitly produced and distributed cannabis, as data collection for this study occurred during 2012–2014, before the decriminalization of medicinal cannabis in Australia in 2016. This is important to note because the cannabinoid and terpene content in illicitly produced cannabis is often unknown to the consumer, and the product itself may contain contaminants, such as pesticides, mold, elevated bacteria levels, and heavy metals that could have potential harmful health effects [37–41]. As such, it is possible that the lack of medical benefits observed in Campbell et al. [15] may be related to the use of unregulated, nonmedical cannabis, rather than a lack of benefits of medical cannabis per se. Moreover, it is important that MC for therapeutic purposes be used under the guidance and supervision of a physician [42]. Although cannabis has a high safety index [42, 43], there are contraindications and precautions that must be taken into consideration for vulnerable populations, such as in those with a history of substance abuse, cardiovascular disease, respiratory disease, and/or anxiety disorders, among other conditions, as well as in patients taking medications that could lead to dangerous drug–drug interactions [44]. Additionally, preclinical studies have found that CBD, as well as co-administered CBD/THC, has biphasic effects, meaning that there is an optimal dose below and above which these cannabinoids are not as effective [45, 46]. More research needs to be conducted to better elucidate the complex mechanisms, as well as the pharmacodynamic and pharmacokinetic properties of cannabinoids, particularly in human populations. Nevertheless, taken together, the precautionary measures that need to be taken into consideration in vulnerable populations and the evidence on biphasic effects of cannabinoids indicate that MC for the purpose of pain management should be used under the care and supervision of a physician, with regular follow-ups to optimize the dose and to monitor issues of tolerability and adverse events.

Notably, our study included subjects who had been cannabis users before initiating MC treatment (light, moderate, and frequent users). As the data collection for the present study took place before the legalization of recreational cannabis in Canada in October of 2018, it is presumed that at least some of the study subjects who reported use before initiating MC treatment would have obtained cannabis through illicit channels, for which there was no information about the type of cannabis obtained, THC and CBD content, or the presence of contaminants. Interestingly, we found that the benefits of MC are beyond those of unregulated and illicitly obtained cannabis. This is evidenced by the findings presented in the Supplementary Data, which indicate significant improvements in mean BPI-S and BPI-I scores compared with baseline, but no significant interactions between the effects of treatment and prior cannabis experience/frequency of use on BPI-S and BPI-I scores (F(12,72)=0.78, P = 0.668, and F(12,72)=0.79, P = 0.655, respectively). This suggests that treatment with MC is associated with improvements in pain regardless of previous experience and/or use of nonmedical cannabis. There are a number of factors that could have contributed to this finding. First—and most importantly—study patients followed a personalized MC treatment plan under the guidance of a physician, with regular checkups to achieve an optimal therapeutic dose while minimizing adverse events due to issues of tolerability. Moreover, by virtue of following such a treatment plan, the psychological factor of guilt and stigma associated with obtaining illicit cannabis is presumed to have been absent or reduced, which in turn would have the effect of reducing anxieties about the use of cannabis, thus further contributing to overall clinical improvement. Finally, illicitly obtained cannabis does not undergo the same testing standards as MC, and inconsistencies in THC/CBD content, as well as cases of the contamination with mold, pesticides, elevated levels of potentially harmful bacteria exceeding Health Canada limits, and heavy metals such as lead—all of which can negatively affect overall health, especially in a population with compromised health—have been reported [37–41]. Taken together, these factors further highlight the importance of following an MC treatment plan under the guidance and supervision of a physician.

Pain alleviation is an important outcome of interest when considering the effectiveness of an analgesic treatment. However, chronic pain is not a condition that is limited to the experience of pain itself. Chronic daily pain also impacts mental and physical health, and consequently negatively affects quality of life [47–49]. As such, quality of life is also an important consideration when evaluating the efficacy of pain treatments [47–49]. In our study, the observed improvements in pain appeared to be associated with positive effects on patients’ health-related quality of life, as evidenced by the observed improvements in measures of physical and mental health in the SF-12. In line with what would be expected in chronic pain patients, PCS and MCS scores were low at baseline, with median scores of 30.18 and 41.59, respectively. Significant improvements were observed over time in both PCS and MCS scores. Given that the analyses of these data were conducted on an unbalanced, incomplete block design, it was not possible to conduct post hoc analyses to further investigate exactly at what time points such changes occurred. However, observation of the ranked mean values would suggest that improvements occurred at M3, with further improvements observed at M6 and M12 on both PCS and MCS scores.

Our findings are consistent with Ware et al. [36], Haroutounian et al. [35], and Bellnier et al. [34], who also conducted long-term prospective open-label studies and found improvements in measures of quality of life after initiation of MC treatment. On the other hand, a recent systematic review and analysis on cannabis-based medicines (including pharmaceutical cannabinoids) for chronic neuropathic pain, as well as a meta-analysis of controlled and observational studies of pharmaceutical cannabinoids, found that these compounds were not superior to placebo in effecting improvements in quality of life [50, 51]. It is difficult to draw comparisons between the findings of Mücke et al. [50], Stockings et al. [51], and our own study findings, as there were some key differences, including study populations, study designs, and outcome measures, among others. It should be noted that the studies by Mücke et al. [50] and Stockings et al. [51] included randomized controlled trials that were shorter in duration, with treatment periods ranging from five days to 14 weeks maximum. Given that in our study (as well as in the studies described above), improvements in MCS and PCS scores were only observed starting at the three-month mark, it is possible that the lack of significant improvements reported by Mücke et al. [50] and Stockings et al. [51] may be partly related to the shorter observation periods of the studies included in their analyses.

Opioid Medication Use

Given the cumulative evidence on the potential benefits of MC as a substitute for opioid medications in the management of pain, exploratory analyses were conducted in a subset of patients who reported using opioid medications at baseline. Examination of OMEs over the course of the study indicated statistically significant reductions in daily opioid medication doses, with reductions observed at M3, and further reductions at M6 and M12. Interestingly, this subset of patients also exhibited significant improvements in pain interference and pain severity. It should be noted, however, that these findings are the results of exploratory analyses in a small subset of patients, and the generalizability of results remains limited due to the high rate of patients lost to follow-up at each subsequent visit.

The mechanisms of action by which cannabinoids confer analgesic effects involve a multitude of regulatory physiological processes. The endocannabinoid system appears to work both independently and synergistically with other molecular targets within major endogenous pain circuitry systems [52]. Cannabinoids are postulated to act at least in part through an opioid receptor mechanism, and preclinical studies have consistently shown synergistic analgesic effects between THC and opioids [53]. Moreover, cannabis has been shown to increase dopamine levels in the nucleus accumbens [54–56], and in humans, it has been found to be associated with reductions in opioid withdrawal symptoms [57, 58], which is postulated to facilitate the process of reducing, and in some cases ceasing, the use of opioid medications. Our findings, albeit exploratory in nature and limited to a small subset of patients, are in line with other large-scale studies that also reported reductions in the use of opioid medications after commencing MC treatment [14, 35, 59–61], suggesting that MC may be an effective and safe treatment option and substitute for opioid medications in the management of pain. This is especially relevant in light of the current opioid crisis.

Safety Profile

Whole-plant cannabis is known to have a high safety index—a one-year prospective cohort study reported that MC is associated with mild to moderate nonserious adverse events, and on its own is not associated with serious adverse events [36]. Accordingly, there have been no reported cases of deaths directly related to cannabis overdose, and the most commonly reported adverse events include dry mouth, increased appetite, and “feeling high” [10, 13, 62, 63]. Our findings were in line with these reports—there were no serious adverse events, and the most commonly reported adverse events after initiating MC were dry mouth, increased appetite, and drowsiness. However, the reported frequencies of these symptoms were not significantly higher than baseline.

Interestingly, in our study, the reported frequencies of fatigue, headaches, feelings of anxiety, and nausea decreased compared with baseline. This finding supports the possibility that MC may be helpful in the management of these symptoms. Of course, such reductions in negative symptomatology may also be attributed to reduction or discontinuation of other medications, switching from recreational/illicitly obtained cannabis to MC, relief of symptoms associated with pain, and/or improvements in physical and mental well-being. Taken together, our findings on safety profile further highlight the importance of following an MC treatment program under the guidance and supervision of a physician to maximize medical benefits while minimizing adverse events.

Generalizability of Results

As indicated in Time Point Definition, and perhaps due to the observational nature of this study, each subsequent follow-up time point had fewer report completions, which may have contributed to biased results and limited the generalizability of results. To investigate potential differences between patients who completed all five time point visits and those who did not, additional analyses were conducted (Supplementary Data). These revealed no significant differences in baseline demographic measures such as age, gender, and type and duration of pain between patients who completed all five time point reports and those who did not. In addition, we evaluated the pattern of treatment response as measured by the BPI in the 5-TMP, 4-TMP, 3-TMP, and 2-TMP groups. Significant improvements were observed from baseline to month 1 in all of these patient groups, and such improvements were maintained up to the last completed time point visit in each group. In other words, significant improvements in BPI were maintained up to month 12 in the 5-TMP group, up to month 6 in the 4-TMP group, and up to month 3 in the 3-TMP group. These results serve as robust evidence that MC effects immediate pain alleviation, as significant improvements were observed after one month of MC treatment in all these patient groups, including the 2-TMP group, which had 580 patients in the analysis, thus suggesting generalizability of results.

On the other hand, it is difficult to determine whether the long-term maintenance of such improvements is also generalizable, as there were fewer report completions at each subsequent visit after month 1 due to patients being lost to follow-up. Given that the improvements in BPI scores were comparably maintained up to month 3 in the 3-TMP, 4-TMP, and 5-TMP groups, and up to month 6 in the 4-TMP and 5-TMP groups, it may be inferred that improvements in BPI scores were similarly maintained in patients who did not complete all the time point reports up to the 12-month follow-up. On the other hand, it is also possible that in patients who did not complete the M3, M6, and/or M12 reports, the improvements in pain observed after one month of treatment may have reverted back to baseline pain levels and then worsened further thereafter. Indeed, there have been reports of pain exacerbation associated with cannabis use [33, 64]. However, such reported negative outcomes appear to be related to cannabis dose and frequency of use, rather than the use of medical cannabis per se, and thus may not be applicable to the present study, where patients followed a treatment regimen under the guidance of a physician. For instance, the study by Wallace et al. [33] reported significant increases in pain at higher doses, which the authors attributed to the existence of a therapeutic window or optimal dose for cannabinoids to effect analgesia, a point that is discussed in Pain Management and Quality of Life, and which further highlights the importance of following a treatment regimen under the guidance of a physician. More recently, Boehnke et al. [64] reported that higher frequency of use is associated with higher pain severity and pain interference than light and moderate frequency of use. However, it must be noted that the differences in pain scores, albeit statistically significant, were rather small (mean BPI-I score of 4.2 in light-frequency users compared with 4.7 in high-frequency users; and mean BPI-S score of 5.2 in light-frequency users compared with 5.6 in high-frequency users). Moreover, the higher pain scores observed in high-frequency compared with moderate- or light-frequency users cannot be concluded to stem from cannabis use, as the study by Boehnke et al. [64] was a cross-sectional study without baseline pain scores to serve as comparison at pre- and postinitiation of MC treatment.

Taken together, the results of the analyses discussed herein suggest that the improvements in pain scores observed after one month of treatment are generalizable to a larger chronic pain population, as improvements were seen in a large sample size of 580 patients. Moreover, given that i) such improvements were similarly maintained in the 5-TMP, 4-TMP, and 3-TMP groups and ii) reports of pain exacerbation associated with cannabis appear to be dose-related and the result of the biphasic effects of cannabinoids rather than the use of MC per se, we propose that the maintenance of pain relief observed in this study may also be generalizable to a larger pain population, as long as patients follow a treatment regimen under the supervision of a physician. Nevertheless, the reproducibility of results in the long term must be confirmed in a study with improved strategies to ensure patient retention for data integrity.

Study Limitations

There were a number of limitations associated with this study. This was an open-label study, and there was no control group. Moreover, the heterogeneity in dose and consistency of cannabis regarding THC/CBD ratio and methods of intake used during the study prevented a more in-depth analysis of dose-related effects with respect to THC and CBD content, as well as the synergistic effects of other cannabinoids, flavonoids, and terpenes, which have been postulated to contribute to the analgesic effects of plant-based MC [52]. It should be noted, however, that great variability in strains and dosing effects is an unavoidable aspect of cannabis therapeutics, and as such, there are limitations to the generalizability of the results of studies that utilize predefined doses vs the customization of best evidence-based practices for individual patients [42, 65]. For instance, dose response is strongly influenced by cannabinoid bioavailability, which has a high degree of intra- and interindividual variability stemming from factors that have been excellently summarized in a review by Huestis [66]. These include—but are not limited to—the mode of administration (e.g., bioavailability can vary from 20% to 30% when taken orally, or 10% to 60% via inhalation), behavioral factors (e.g., recent meals, depth of inhalation, duration of breath holding, and temperature of vaporizer can all affect cannabis absorption), and genetic factors (e.g., high intra- and interindividual variability in the activity rates of enzymes involved in cannabinoid metabolism, such as CYP 450 2C9 and 2A), among others [66]. Accordingly, MC doses are individually determined, and the general approach to MC treatment is to “start slow, go slow, and stay low,” increasing the dose as needed and as tolerated [42]. In this regard, medical cannabis is a highly individualized evidence-based medicine, where a high degree of variability in cannabis strain and dosing is to be expected, as the patient serves as the sole unit of observation for efficacy and tolerability in finding the optimal intervention or combination of plant varieties and dosage forms [42].

Another limitation of the study was that, as discussed in Generalizability of Results, each subsequent follow-up time point had fewer report completions, which may have contributed to biased results and limited the generalizability of results. The issue of unbalanced data sets due to patients lost to follow-up is not uncommon, particularly in large-scale, long-term studies [67, 68]. In this study, the issue may have been exacerbated by the lack of financial incentives to complete reports and the fact that prompts to complete follow-ups reports were sent via e-mails that contained links to the questionnaires. While this method may have been beneficial in terms of time- and cost-efficiency, it is also likely to have been disadvantageous in that it lacked the “personal touch” necessary to maintain good rapport and motivation for study participants to complete follow-up study reports [68]. In this regard, the data collection methods and strategies to retain long-term patients were limitations of the study.

Another limitation of the study was the lack of standardized reporting for adverse events associated with MC treatment. As this was an observational study, we tried to keep the time burden of completing questionnaires low. As such, several aspects of adverse event data collection common in clinical trials (e.g., duration, severity, action[s] taken with the intervention, rescue medications, outcome) were not collected in this study, which prevented more in-depth analyses.

Finally, other limitations of the study include selection/volunteer bias, as this was a study of patients at a medical cannabis clinic who had chosen MC to treat their chronic pain and thus was not a full representation of the chronic pain population at large, and recall and expectancy bias, as data in the study were collected by means of subjective self-report questionnaires.

Conclusions

In this large, open-label, prospective, observational study, we found that treatment with MC was associated with significant improvements in measures of pain as well as quality of life, which were sustained over the course of the 12-month observation period in patients who remained in the study for the full duration. Improvements in measures of pain were observed as early as one month after initiating treatment with MC, while improvements in quality of life were only observed starting at three months after initiating treatment. This suggests that while the analgesic effects have an immediate onset of action, the global health benefits of MC necessitate a longer treatment course. We also found that MC was associated with significant improvements in measures of pain regardless of previous recreational use, indicating that the benefits of an MC treatment course extend beyond those of unregulated, recreational cannabis. Such benefits are postulated to stem from the facts that medical-grade cannabis meets higher standards of purity and consistency and that there are no contaminants such as mold, pesticides, or other substances that could negatively affect health. Additionally, following a treatment course that is tailored to the needs and tolerability of the patient, under the guidance of a physician, is likely to contribute to acceptance and treatment adherence by the patient, which in turn may have further contributed to improved pain scores and quality of life. Moreover, the MC treatment course in this study was not associated with increases in the frequency of undesired adverse events, but rather decreased the frequency of headaches, fatigue, feelings of anxiety, and nausea, further supporting the importance of following a treatment course that is optimized to the needs and tolerability of the patient under the supervision of a physician. Finally, in a small subset of patients who reported using opioid medications at baseline, exploratory analyses revealed that opioid medication doses decreased, while measures of pain were significantly improved over the course of the study treatment observation period. Taken together, the results of this study add to the cumulative evidence in support of plant-based MC as a safe and effective treatment option and potential opioid substitute or augmentation therapy for the management of chronic pain symptomatology and quality of life.

Funding sources: This study was funded by Apollo Applied Research.

Disclosure and conflicts of interest: Authors Vahid Salimpour and SunYoung Rosalia Yoon were affiliated with and Bryan Hendin and Genane Loheswaran were employees of Apollo Applied Research during the time the study was conducted and/or the manuscript was written. Apollo Applied Research is an independent subsidiary of Canopy Growth Corp. Author Gordon Ko was affiliated with Apollo Applied Research at the time the study was conducted/the manuscript was written, and he has received CME honoraria or support, consultancy fees, or grants from the following companies during 2016–2019: Aurora (CanniMed, MedReleaf), Canopy (Mettrum, Tweed, Apollo), Canntrust/Tetra BioPharma; Tilray (CBM talks/clinical advisory boards participation). Authors Ramin Safakish and Imrat Sohanpal have no conflicts of interest to declare.

Acknowledgments

We would like to thank Dr. Paula Williams and Bijan Rafat for their intellectual contributions to study design and data collection.

Supplementary Data

Supplementary data are available at Pain Medicine online.

References

1

Breivik
H
,
Collett
B
,
Ventafridda
V
,
Cohen
R
,
Gallacher
D.
 
Survey of chronic pain in Europe: Prevalence, impact on daily life, and treatment
.
Eur J Pain
 
2006
;
10
(
4
):
287
333
.

2

Johannes
CB
,
Le
TK
,
Zhou
X
,
Johnston
JA
,
Dworkin
RH.
 
The prevalence of chronic pain in United States adults: Results of an Internet-based survey
.
J Pain
 
2010
;
11
(
11
):
1230
9
.

3

Pitcher
MH
,
von Korff
M
,
Bushnell
MC
,
Porter
L.
 
Prevalence and profile of high-impact chronic pain in the United States
.
J Pain
 
2019
;
20
(
2
):
146
60
.

4

Goldberg
DS
,
McGee
SJ.
 
Pain as a global public health priority
.
BMC Public Health
 
2011
;
11
(
1
):
770
.

5

Schopflocher
D
,
Taenzer
P
,
Jovey
R.
 
The prevalence of chronic pain in Canada
.
Pain Res Manag
 
2011
;
16
(
6
):
445
50
.

6

Gaskin
DJ
,
Richard
P.
 
The economic costs of pain in the United States
.
J Pain
 
2012
;
13
(
8
):
715
24
.

7

Jones
CM
,
Mack
KA
,
Paulozzi
LJ.
 
Pharmaceutical overdose deaths, United States, 2010
.
JAMA
 
2013
;
309
(
7
):
657
9
.

8

Kalso
E
,
Edwards
JE
,
Moore
RA
,
McQuay
HJ.
 
Opioids in chronic non-cancer pain: Systematic review of efficacy and safety
.
Pain
 
2004
;
112
(
3
):
372
80
.

9

Noble
M
,
Treadwell
JR
,
Tregear
SJ
, et al. .
Long-term opioid management for chronic noncancer pain
.
Cochrane Database Syst Rev
 
2010
;
1
:
CD006605
.

10

Rudd
RA
,
Aleshire
N
,
Zibbel
JE
,
Gladden
RM.
 
Increases in drug and opioid overdose deaths—United States, 2000-2014
.
MMWR Morb Mortal Wkly Rep
 
2016
;
64
(
50–51
):
1378
82
.

11

Hedegaard
H
,
Warmer
M
,
Minino
AM.
 
Drug overdose deaths in the United States, 1999-2016
.
NCHS Data Brief
 
2017
;
294
:
1
8
.

12

Bachhuber
MA
,
Saloner
B
,
Cunningham
CO
,
Barry
CL.
 
Medical cannabis laws and opioid analgesic overdose mortality in the United States, 1999-2010
.
JAMA Intern Med
 
2014
;
174
(
10
):
1668
73
.

13

Vyas
MB
,
LeBaron
VT
,
Gilson
AM.
 
The use of cannabis in response to the opioid crisis: A review of the literature
.
Nurs Outlook
 
2018
;
66
(
1
):
56
65
.

14

Boehnke
KF
,
Scott
JR
,
Litinas
E
,
Sisley
S
,
Williams
DA
,
Clauw
DJ.
 
Pills to pot: Observational analyses of cannabis substitution among medical cannabis users with chronic pain
.
J Pain
 
2019
;
20
(
7
):
830
41
.

15

Campbell
G
,
Hall
WD
,
Peacock
A
, et al. .
Effect of cannabinoid use in people with chronic non-cancer pain prescribed opioids: Findings from a 4-year prospective cohort study
.
Lancet Public Health
 
2018
;
3
(
7
):
e341
50
.

16

Cleeland
CS
,
Ryan
KM.
 
Pain assessment: Global use of the Brief Pain Inventory
.
Ann Acad Med Singapore
 
1994
;
23
(
2
):
129
38
.

17

Gjeilo
KH
,
Stenseth
R
,
Wahba
A
,
Lydersen
S
,
Klepstad
P.
 
Validation of the Brief Pain Inventory in patients six months after cardiac surgery
.
J Pain Symptom Manage
 
2007
;
34
(
6
):
648
56
.

18

Jelsness-Jørgensen
LP
,
Mourn
B
,
Grimstad
T
, et al. .
Validity, reliability, and responsiveness of the Brief Pain Inventory in inflammatory bowel disease
.
Can J Gatroenterol Hepatol
 
2016
;
2016
:
5624261
.

19

Ware
JE
,
Sherbourne
CS.
 
The MOS 36-item Short-Form Health Survey (SF-36). I. Conceptual framework and item selection
.
Med Care
 
1992
;
30
(
6
):
473
83
.

20

Ware
J
,
Kosinski
M
,
Keller
SD.
 
SF-12: How to Score the SF-12 Physical and Mental Health Summary Scales
. 2nd ed.
Boston
:
Health Institute, New England Medical Center
;
1995
.

21

Resnick
B
,
Parker
R.
 
Simplified scoring and psychometrics of the revised 12-item Short-Form Health Survey
.
Outcomes Manag Nurs Pract
 
2001
;
5
(
4
):
161
6
.

22

Nielsen
S
,
Degenhardt
L
,
Hoban
B
,
Gisev
N.
 
A synthesis of oral morphine equivalents (OME) for opioid utilisation studies
.
Pharmacoepidemiol Drug Saf
 
2016
;
25
(
6
):
733
7
.

23

Chatfield
M
,
Mander
A.
 
The Skillings-Mack test (Friedman test when there are missing data)
.
Stata J
 
2009
;
9
(
2
):
299
305
.

24

Laerd Statistics. Cochran’s Q test using SPSS Statistics. Statistical tutorial and software guides.

2017
. Available at: https://statistics.laerd.com/ (accessed August 2019).

25

Serlin
RC
,
Mendoza
TR
,
Nakamura
Y
,
Edwards
KR
,
Cleeland
CS.
 
When is cancer pain mild, moderate or severe? Grading pain severity by its interference with function
.
Pain
 
1995
;
61
:
277
84
.

26

Boonstra
AM
,
Preuper
HRS
,
Balk
GA
,
Stewart
RE.
 
Cut-off points for mild, moderate, and severe pain on the visual analogue scale for pain in patients with chronic musculoskeletal pain
.
Pain
 
2014
;
155
(
12
):
2545
50
.

27

Farrar
JT
,
Young
JP
Jr
,
LaMoreaux
L
,
Werth
JL
,
Poole
RM.
 
Clinical importance of changes in chronic pain intensity measured on an 11-point numerical pain rating scale
.
Pain
 
2001
;
94
(
2
):
149
58
.

28

Abrams
DI
,
Jay
CA
,
Shade
SB
, et al. .
Cannabis in painful HIV-associated sensory neuropathy: A randomized placebo-controlled trial
.
Neurology
 
2007
;
68
(
7
):
515
21
.

29

Ellis
RJ
,
Toperoff
W
,
Vaida
F
, et al. .
Smoked medical cannabis for neuropathic pain in HIV: A randomized, crossover clinical trial
.
Neuropsychopharmacology
 
2009
;
34
(
3
):
672
80
.

30

Ware
MA
,
Wang
T
,
Shapiro
S
, et al. .
Smoked cannabis for chronic neuropathic pain: A randomized controlled trial
.
CMAJ
 
2010
;
182
(
14
):
E694
701
.

31

Wilsey
B
,
Marcotte
T
,
Deutsch
R
,
Gouaux
B
,
Sakai
S
,
Donaghe
H.
 
Low-dose vaporized cannabis significantly improves neuropathic pain
.
J Pain
 
2013
;
14
(
2
):
136
48
.

32

Wallace
MS
,
Marcotte
TD
,
Umlauf
A
,
Gouaux
B
,
Atkinson
JH.
 
Efficacy of inhaled cannabis on painful diabetic neuropathy
.
J Pain
 
2015
;
16
(
7
):
616
27
.

33

Wallace
M
,
Schulteis
G
,
Atkinson
JH
, et al. .
Dose-dependent effects of smoked cannabis on capsaicin-induced pain and hyperalgesia in healthy volunteers
.
Anesthesiology
 
2007
;
107
(
5
):
785
96
.

34

Bellnier
T
,
Brown
GW
,
Ortega
TR.
 
Preliminary evaluation of the efficacy, safety, and costs associated with the treatment of chronic pain with medical cannabis
.
Ment Health Clin
 
2018
;
8
(
3
):
110
5
.

35

Haroutounian
S
,
Ratz
Y
,
Ginosar
Y
, et al. .
The effect of medicinal cannabis on pain and quality of life outcomes in chronic pain: A prospective open-label study
.
Clin J Pain
 
2016
;
32
(
12
):
1036
43
.

36

Ware
MA
,
Wang
T
,
Shapiro
S
, et al. .
COMPASS study team. Cannabis for the management of pain: Assessment of safety study (COMPASS)
.
J Pain
 
2015
;
16
(
12
):
1233
42
.

37

Busse
FP
,
Fiedler
GM
,
Leichtle
A
,
Hentschel
H
,
Stumvoll
M.
 
Lead poisoning due to adulterated marijuana in Leipzig
.
Dtsch Arztebl Int
 
2008
;
105
(
44
):
757
2
.

38

McLaren
J
,
Swift
W
,
Dillon
P
,
Allsop
S.
 
Cannabis potency and contamination: A review of the literature
.
Addiction
 
2008
;
103
(
7
):
1100
9
.

39

Schneider
S
,
Bebing
R
,
Dauberschmidt
C.
 
Detection of pesticides in seized illegal cannabis plants
.
Anal Methods
 
2014
;
6
(
2
):
515
20
.

40

Robertson
G
,
McArthur
G.
Globe investigation: What’s in your weed? We tested dispensary marijuana to find out. The Globe and Mail. July 27, 2016. Available at: https://www.theglobeandmail.com/cannabis/article-globe-investigation-whats-in-your-weed-we-tested-dispensary/ (accessed January 2020).

41

Owram
K.
It’s caveat emptor for online CBD shoppers. Bloomberg. Available at: https://www.bloomberg.com/news/articles/2019-10-20/it-s-caveat-emptor-for-online-cbd-shoppers-cannabis-weekly (accessed January
2020
).

42

MacCallum
CA
,
Russo
EB.
 
Practical considerations in medical cannabis administration and dosing
.
Eur J Intern Med
 
2018
;
49
:
12
9
.

43

Herkenham
M
,
Lynn
AB
,
Little
MD
, et al. .
Cannabinoid receptor location in brain
.
Proc Natl Acad Sci U S A
 
1990
;
87
(
5
):
1932
6
.

44

Kahan
M
,
Srivastava
A
,
Spithoff
S
,
Bromley
L.
 
Prescribing smoked cannabis for chronic noncancer pain: Preliminary recommendations
.
Can Fam Physician
 
2014
;
60
(
12
):
1083
90
.

45

Malfait
AM
,
Gallily
R
,
Sumariwalla
PF
, et al. .
The nonpsychoactive cannabis constituent cannabidiol is an oral anti-arthritic therapeutic in murine collagen-induced arthritis
.
Proc Natl Acad Sci U S A
 
2000
;
97
(
17
):
9561
6
.

46

Casey
SL
,
Atwal
N
,
Vaughan
CW.
 
Cannabis constituent synergy in a mouse neuropathic pain model
.
Pain
 
2017
;
158
:
2452
60
.

47

Dansie
EJ
,
Turk
DC.
 
Assessment of patients with chronic pain
.
Br J Anaesth
 
2013
;
111
(
1
):
19
25
.

48

Turk
DC
,
Dworkin
RH
,
Revicki
D
, et al. .
Identifying important outcome domains for chronic pain clinical trials: An IMMPACT survey of people with pain
.
Pain
 
2008
;
137
(
2
):
276
85
.

49

Salaffi
F
,
Sarzi-Puttini
P
,
Ciapetti
A
,
Atzeni
F.
 
Assessment instruments for patients with fibromyalgia: Properties, applications and interpretation
.
Clin Exp Rheumatol
 
2009
;
27
(5 Suppl 56):
S92
105
.

50

Mücke
M
,
Phillips
T
,
Radbruch
L
,
Petzke
F
,
Hauser
W.
 
Cannabis-based medicines for chronic neuropathic pain in adults
.
Cochrane Database Syst Rev
 
2018
;
3
:
CD012182
.

51

Stockings
E
,
Campbell
G
,
Hall
WD
, et al. .
Cannabis and cannabinoids for the treatment of people with chronic noncancer pain conditions: A systematic review and meta-analysis of controlled and observational studies
.
Pain
 
2018
;
159
(
10
):
1932
54
.

52

Baron
EP.
 
Medicinal properties of cannabinoids, terpenes, and flavonoids in cannabis, and benefits in migraine, headache, and pain: An update on current evidence and cannabis science
.
Headache
 
2018
;
58
(
7
):
1139
86
.

53

Nielsen
S
,
Sabioni
P
,
Trigo
JM
, et al. .
Opioid-sparing effect of cannabinoids: A systematic review and meta-analysis
.
Neuropsychopharmacology
 
2017
;
42
(
9
):
1752
65
.

54

Tanda
G
,
Pontieri
FE
,
di Chiara
G.
 
Cannabinoid and heroin activation of mesolimbic dopamine transmission by a common μ1 opioid receptor mechanism
.
Science
 
1997
;
276
(
5321
):
2048
50
.

55

Scavone
JL
,
Sterling
RC
,
van Bockstaele
EJ.
 
Cannabinoid and opioid interactions: Implications for opiate dependence and withdrawal
.
Neuroscience
 
2013
;
248
:
637
54
.

56

Ashton
CH.
 
Pharmacology and effects of cannabis: A brief review
.
Br J Psychiatry
 
2001
;
178
(
2
):
101
6
.

57

Hermann
D
,
Klages
E
,
Welzel
H
,
Mann
K
,
Croissant
B.
 
Low efficacy of non-opioid drugs in opioid withdrawal symptoms
.
Addict Biol
 
2005
;
10
(
2
):
165
9
.

58

Scavone
JL
,
Sterling
RC
,
Weinstein
SP
,
van Bockstaele
EJ.
 
Impact of cannabis use during stabilization on methadone maintenance treatment
.
Am J Addict
 
2013
;
22
(
4
):
344
51
.

59

Lucas
P
,
Walsh
Z.
 
Medical cannabis access, use, and substitution for prescription opioids and other substances: A survey of authorized medical cannabis patients
.
Int J Drug Policy
 
2017
;
42
:
30
5
.

60

Abuhasira
R
,
Schleider
LB
,
Mechoulam
R
,
Novack
V.
 
Epidemiological characteristics, safety and efficacy of medical cannabis in the elderly
.
Eur J Intern Med
 
2018
;
49
:
44
50
.

61

Bar-Lev Schleider
L
,
Mechoulam
R
,
Lederman
V
, et al. .
Prospective analysis of safety and efficacy of medical cannabis in large unselected population of patients with cancer
.
Eur J Intern Med
 
2018
;
49
:
37
43
.

62

United States Department of Health and Human Services (US DHHS), Centers for Disease Control and Prevention (CDC), National Center for Health Statistics (NCHS).

Multiple Causes of Death 1999–2014
.
Atlanta
:
Centers for Disease Control
;
2015
.

63

Aviram
J
,
Samuelly-Leichtag
G.
 
Efficacy of cannabis-based medicines for pain management: A systematic review of meta-analysis of randomized controlled trials
.
Pain Physician
 
2017
;
20
(
6
):
E755
96
.

64

Boehnke
KF
,
Scott
JR
,
Litinas
E
,
Sisley
S
,
Williams
DA
,
Clauw
DJ.
 
High-frequency medical cannabis use is associated with worse pain among individuals with chronic pain
.
J Pain
. In press.

65

Frieden
TR.
 
Evidence for health decision making—beyond randomized, controlled trials
.
N Engl J Med
 
2017
;
377
(
5
):
465
75
.

66

Huestis
MA.
 
Human cannabinoid pharmacokinetics
.
Chem Biodivers
 
2007
;
4
(
8
):
1770
804
.

67

Kaur
M
,
Sprague
S
,
Ignacy
T
,
Thoma
A
,
Bhandari
M
,
Farrokhyar
F.
 
How to optimize participant retention and complete follow-up in surgical research
.
Can J Surg
 
2014
;
57
(
6
):
420
7
.

68

Tong
SC
,
Tin
AS
,
Lim
JFY
,
Chow
WL.
 
Innovative proven clinical-research strategies for participant recruitment and retention
.
Proc Singapore Healthc
 
2010
;
19
(
1
):
64
8
.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://academic.oup.com/journals/pages/open_access/funder_policies/chorus/standard_publication_model)

Supplementary data