Vaporized Cannabis versus Placebo for Acute Migraine: A Randomized Controlled Trial (2024)

As a library, NLM provides access to scientific literature. Inclusion in an NLM database does not imply endorsem*nt of, or agreement with, the contents by NLM or the National Institutes of Health.
Learn more: PMC Disclaimer | PMC Copyright Notice

Vaporized Cannabis versus Placebo for Acute Migraine: A Randomized Controlled Trial (1)

Link to Publisher's site

Version 1. medRxiv. Preprint. 2024 Feb 18.

PMCID: PMC10889030

PMID: 38405890

This is a preprint.

It has not yet been peer reviewed by a journal.

The National Library of Medicine is running a pilot to include preprints that result from research funded by NIH in PMC and PubMed.

Author information Copyright and License information PMC Disclaimer

The complete version history of this preprint is available at medRxiv.

Abstract

Background

Preclinical and retrospective studies suggest cannabinoids may be effective in migraine treatment. However, there have been no randomized clinical trials examining the efficacy of cannabinoids for acute migraine.

Methods

In this randomized, double-blind, placebo-controlled, crossover trial, adults with migraine treated up to 4 separate migraine attacks, 1 each with vaporized 1) 6% Δ9-tetrahydrocannabinol (THC-dominant); 2) 11% cannabidiol (CBD-dominant); 3) 6% THC+11% CBD; and 4) placebo cannabis flower in a randomized order. Washout period between treated attack was ≥1 week. The primary endpoint was pain relief and secondary endpoints were pain freedom and most bothersome symptom (MBS) freedom, all assessed at 2 hours post-vaporization.

Results

Ninety-two participants were enrolled and randomized, and 247 migraine attacks were treated. THC+CBD was superior to placebo at achieving pain relief (67.2% vs 46.6%, Odds Ratio [95% Confidence Interval] 2.85 [1.22, 6.65], p=0.016), pain freedom (34.5% vs. 15.5%, 3.30 [1.24, 8.80], p=0.017) and MBS freedom (60.3% vs. 34.5%, 3.32 [1.45, 7.64], p=0.005) at 2 hours, as well as sustained pain freedom at 24 hours and sustained MBS freedom at 24 and 48 hours. THC-dominant was superior to placebo for pain relief (68.9% vs. 46.6%, 3.14 [1.35, 7.30], p=0.008) but not pain freedom or MBS freedom at 2 hours. CBD-dominant was not superior to placebo for pain relief, pain freedom or MBS freedom at 2 hours. There were no serious adverse events.

Conclusions

Acute migraine treatment with 6% THC+11% CBD was superior to placebo at 2 hours post-treatment with sustained benefits at 24 and 48 hours.

Introduction

Migraine is the second leading cause of years lived with disability worldwide. It affects over a billion people worldwide, including 38 million Americans1. Migraine treatments are classified for acute and/or preventive use2. Nearly everyone with migraine uses acute treatments for migraine attacks. However, rates of treatment optimization with traditional acute therapies are relatively low and rates of discontinuation are high3,4. There is significant interest in and use of cannabinoids for acute migraine treatment. Migraine is among the most common medicinal uses of cannabinoids, with 35.5% of 1,429 medical cannabis users reporting use for headache/migraine; inhalation was the most common route of administration (81.4%)5.

More than 125 identified phytocannabinoids are naturally found in the cannabis plant, including Δ9-tetrahydrocannabinol (THC) and cannabidiol (CBD). Although the US Food and Drug Administration (FDA) has approved cannabinoid-based medications for specific conditions, none have been approved for migraine. At the time of this writing, THC was legal for medical and/or recreational use in 38 US states and the District of Columbia. Since 2018, hemp-derived CBD (<0.3% THC) is not a controlled substance in the US6.

Patients often ask healthcare professionals about cannabinoids for migraine, but data to inform medical advice is lacking7. Preclinical evidence suggests that cannabinoids may have effects on migraine pathogenesis through mechanisms including inhibiting calcitonin gene-related peptide (CGRP) release, inhibiting CGRP-induced nitric oxide release, inhibiting trigeminovascular neurons, and inhibiting cortical spreading depression, and surveys and retrospective studies suggest that cannabinoids may have benefit as acute and/or preventive treatments for migraine820. However the efficacy of cannabis for acute treatment of migraine has not been studied via a randomized, controlled trial (RCT)10,12,21. Therefore, this RCT was conducted to assess the efficacy of vaporized cannabis against placebo cannabis.

Methods

Trial Design

This randomized, double-blind, placebo-controlled, crossover trial was conducted at the University of California, San Diego (UCSD). The study was approved by the UCSD Institutional Review Board (IRB #181944) and the US FDA. The study was prospectively registered, and full protocol is available, at ClinicalTrials.gov (NCT04360044). The authors take full responsibility for the data and the accuracy and integrity of the publication. No writing assistance was provided by outside parties. The study was performed in accordance with the principles of the Declaration of Helsinki. All participants provided written informed consent prior to enrollment.

Sample Size

Sample size was determined based on our primary outcome of pain relief at 2 hours, assuming a response of 68% for treatment and 45% for placebo as seen in an intranasal sumatriptan RCT22. This effect size required 72 participants to detect a significant difference with 80%. Assuming a 20% dropout rate, we planned to enroll 90 participants.

Study Drug

Cannabis flower was obtained from the National Institute on Drug Abuse (NIDA) Drug Supply Program (DSP) and consisted of 4 different treatments. The most closely matched batches available from the NIDA DSP at study initiation were selected. They were: 1) 5.62% THC+0.03% CBD (referred to in this study as 6% THC or THC-dominant); 2) 11.27% CBD+0.35% CBD (referred to as 11% CBD or CBD-dominant); 3) 6.16% THC + 10.77% CBD (referred to as 6% THC+11% CBD or THC+CBD); and 4) placebo flower (<0.025% THC + 0.14% CBD). All treatments contained <1% minor cannabinoids and were devoid of terpenes. The studied THC potency was based on previous pain studies2325.

Participants

Key inclusion criteria were: ages 21–65; migraine according to International Classification of Headache Disorders, 3rd edition (ICHD-3) criteria26; 2–23 headache days and 2–23 migraine days per month; agree not to use cannabis (outside of the study drug), opioids or barbiturates. Key exclusion criteria were: screening visit urine drug test positive for THC, barbiturates, opioids, oxycodone, or methadone; pregnant; breastfeeding; known cognitive impairment; current moderate-severe or severe depression; history of bipolar disorder, schizophrenia, or psychosis; history of substance use disorder; and active pulmonary disease or other severe medical illnesses at the discretion of the researchers. Inclusion criteria initially required THC use within the prior 2 years; this was removed March 18, 2021 due to slow recruitment and no serious adverse events at that time. Full criteria are available at ClinicalTrials.gov.

Participants agreed not to use any other acute migraine treatments prior to or within 2 hours following study drug administration.

Enrollment

Participants were enrolled at UCSD November 20, 2020-November 4, 2022 with follow-up completed February 23, 2023. A headache neurologist experienced in ICHD-3 criteria (NMS) confirmed participant eligibility.

After written informed consent, baseline characteristics were captured using REDCap27. Then participants were trained in the Foltin Uniform Puff Procedure (FUPP), a validated cannabis vaporization procedure, and a smartphone application was installed on participants’ smartphones to guide participants through the study and collect data28.

Randomization

A research pharmacist randomized participants to receive the 4 different treatments using simple 1:1:1:1 assignment (24 possible orders) using Microsoft Excel’s random number function.

Blinding

Participants, research coordinators, investigators, and statisticians were blinded until after the initial statistical analysis was completed; only the research pharmacists were unblinded during the study. The four treatments were prepared into identical Storz & Bickel Filling Set vaporization capsules by a research pharmacist and placed into sealed bags. Labels were affixed to the sealed bags stating the order in which the participant would use the four treatments (“Migraine 1” through “Migraine 4”). The key linking the treatments to their identifying number was stored on a password-protected computer in the locked pharmacy available only to the research pharmacists.

Participant blinding was promoted by using placebo cannabis flower from which the THC and CBD had been extracted via a chemical process by the NIDA DSP and by framing, including educating patients that they might experience a “placebo high” from the CBD-dominant and placebo treatments and that they might not experience a “high” from the THC-dominant and THC+CBD treatments as the study potencies were lower than “recreational” potencies. Each attack, after answering 2-hour efficacy and safety questions, participants were asked which treatment they thought they received.

Treatments

Upon each migraine attack onset, the participant accessed the smartphone application. If ≥7 days had elapsed since the prior cannabis administration (ensuring ≥7 days washout period between cannabis administrations) the application asked the participant questions to establish whether the attack met treatment criteria of: 1) headache <4 hours from onset, 2) headache pain moderate or severe in intensity, 3) associated with photophobia and phonophobia and/or with nausea, 4) no acute treatments used since attack onset. If the attack met all criteria, the application instructed the participant to vaporize study cannabis at 180° Celsius and guided the participant through the timed FUPP to standardize vaporization procedure across treated attacks and across participants. FUPP consists of five seconds of inhalation, followed by a ten second breath hold, exhalation, and 45 second waiting period before repeating the process28. Each attack, participants performed the FUPP 4 times under continuous application guidance. The application pushed surveys at 1, 2, 24 and 48 hours.

Outcomes

Outcomes were based on International Headache Society (IHS) guidelines29. Primary outcome was pain relief at 2 hours post-vaporization29 based on the limited supporting evidence at study initiation18. Secondary outcomes were 2-hour pain freedom and 2-hour most bothersome symptom (MBS) freedom29. Other outcomes included freedom from pain, photophobia, phonophobia, nausea, and vomiting, and rescue medication use, with data collected at 1, 2, 24 and 48 hours29. Pain at time 0 and each subsequent timepoint was reported as none, mild, moderate or severe29. Pain relief was defined as reduction of pain from moderate or severe to mild or none29. Pain freedom was defined as absence of pain. Sustained pain freedom was defined as pain freedom at 2 hours without return of pain or rescue medication use29. MBS (selected from photophobia, phonophobia, or nausea) was identified by the participant during each attack prior to vaporization; MBS freedom was defined as absence of the MBS at each subsequent timepoint. Sustained MBS freedom was defined as MBS freedom at 2 hours without return of the MBS or rescue medication use at each subsequent timepoint 29. All outcomes presented here were prospectively registered (NCT04360044).

Analyses

The intention-to-treat (ITT) analysis analyzed all treated migraine attacks, counting treated attacks without recorded 2-hour data as treatment failures as per IHS guidelines29. The modified intention-to-treat analysis (mITT) only analyzed attacks with recorded 2-hour data. During the trial, some subjects filled out surveys early or late. We performed a sensitivity analysis analyzing only attacks with 2-hour data time stamped 1.5–3 hours post-vaporization, a time window selected to retain as much data as possible without increasing possible exposure to retention bias3032. As per IHS guidelines, for all analyses, attacks for which the participant used rescue medication before the 2-hour assessment were counted as treatment failures29. The differences in pain relief, pain freedom, and MBS freedom among the four treatments were assessed using a generalized linear mixed effects model with a logit link for binary outcomes. A random intercept structure was included to account for the cluster effect of subjects going through the same trial multiple times. Secondary and other analyses at 1, 24, and 48 hours were conducted using similar methods. Statistical analyses were performed using RStudio, version 4.1.2 (R Foundation). All tests were two-sided with p<0.05 indicating statistical significance.

Results

678 people were screened for eligibility, of whom 92 were enrolled (Figure 1). Participants had a median age of 41, and 82.6% were female (Table 1).

Vaporized Cannabis versus Placebo for Acute Migraine: A Randomized Controlled Trial (2)

Enrollment, Randomization and Follow-up

Table 1:

Characteristics of Patient Population

CharacteristicAll Patients (n=92)
Age-median years (Quartiles 1–3)41 (34–54)
Birth Sex- no. (%)
 Female76 (82.6%)
 Male16 (17.4%)
Gender Identity-no. (%)
 Female sex77 (83.7%)
 Male sex15 (16.3%)
BMI-median (Quartiles 1–3)26.0 (22.8–32.9)
Ethnicity
 Hispanic or Latino15 (16.3%)
 Not Hispanic or Latino77 (83.7%)
Race or ethnic group-no.(%)
 White71 (77.2%)
 Black8 (8.9%)
 Asian-American7 (7.8%)
 Mixed/Other2 (2.2%)
 Middle Eastern1 (1.1%)
 Pacific Islander1 (1.1%)
 Declined to state2 (2.2%)
Primary language- no. (%)
 English83 (90.2%)
 Spanish6 (6.5%)
 Other3 (3.3%)
Migraine history
 Age at onset- median years (Quartiles 1–3)20 (14–28)
 Headache Days per month- median (Quartiles 1–3)15 (8–16)
 Migraine Days per month- median (Quartiles 1–3)6 (4–10)
 Chronic Migraine*- no. (%)25/91 (27.5%)
Most bothersome symptom, treated attack
 Photophobia148 (63.2%)
 Phonophobia38 (16.2%)
 Nausea48 (20.5%)
Last Cannabis Use
 Never33 (35.9%)
 More than 1 year aqo19 (20.7%)
 6 months-1 year ago7 (7.6%)
 1 month-6 months ago19 (20.7%)
 2 weeks-1 month ago5 (5.4%)
 Last 2 weeks4 (4.3%)
 Incomplete Data5 (5.4%)

*Chronic Migraine: At least 15 headache days and at least 8 migraine days per month

The ITT analysis included all 247 migraine attacks treated with vaporized cannabis from 73 participants. The mITT analysis included 234 attacks from 71 patients. The sensitivity analysis included 202 attacks from 70 patients (Figure 1). The results were similar across the ITT, mITT, and sensitivity analyses for the primary endpoint of 2-hour pain relief and secondary endpoints of 2-hour pain freedom and 2-hour MBS freedom. Where not otherwise specified, mITT results are reported as they neither used imputed data (as in the ITT) nor excluded data (as in the sensitivity analysis).

For the primary outcome of 2-hour pain relief, THC+CBD and THC-dominant were superior to placebo but CBD-dominant was not in all 3 analyses. In the ITT analysis, 2-hour pain relief responder rates were 63.9% with THC+CBD, 67.7% with THC-dominant, 50% with CBD-dominant, and 42.2% with placebo. In the mITT analysis, 2-hour pain relief was achieved by 67.2% with THC+CBD (OR [95%] 2.85 [1.22–6.65], p=0.016), 68.9% with THC-dominant (3.140 [1.35–7.30], p=0.008), 52.6% with CBD-dominant (p>0.05), and 46.6% with placebo (Figures 2 and and3).3). In the sensitivity analysis; 2-hour pain relief responder rates were 64.0% with THC+CBD, 70.0% with THC-dominant, 53.1% with CBD-dominant, and 45.3% with placebo.

Vaporized Cannabis versus Placebo for Acute Migraine: A Randomized Controlled Trial (3)

Box Plots of Efficacy Data

2A) Efficacy Outcomes at 2 Hours. P-values were based on generalized linear mixed effects model.

2B) Pain Freedom rates at 1h and 2h and Sustained Pain Freedom rates at 24h and 48h

2C) Most Bothersome Symptom (MBS) Freedom rates at 1h and 2h and Sustained MBS Freedom rates at 24h and 48h (* p<0.05)

Vaporized Cannabis versus Placebo for Acute Migraine: A Randomized Controlled Trial (4)

Forest Plots of Efficacy Data

For the secondary outcomes, THC+CBD was superior to placebo in all 3 analyses for 2-hour pain freedom (mITT 34.5% vs. 15.5%, 3.30 [1.24–8.80], p=0.017) and 2-hour MBS freedom (mITT 60.3% vs. 34.5%, 3.32 [1.45–7.64], p=0.005) but THC-dominant and CBD-dominant were not (Figures 2 and and3).3). Adjusting for treatment session number did not affect the results of the primary or secondary outcomes.

THC+CBD was superior to placebo at 2 hours with regards to freedom from photophobia and phonophobia but not nausea or vomiting, while THC-dominant and CBD-dominant were not superior to placebo for any of these (Figure 3).

At 1 hour, pain relief responder rate for THC+CBD (53.6%, 2.56 [1.05–6.25], p=0.039), THC-dominant (65.5%, 4.72 [1.89–11.81], p<0.001), and CBD-dominant (58.9%, 3.20 [1.31–7.82], p=0.011) were all superior to 36.7% responder rate with placebo. 1-hour pain freedom was superior only for THC-dominant vs. placebo (17.2% vs 5.0%, 4.90 [1.12–21.34], p=0.034), and 1-hour MBS freedom was superior for THC-dominant (37.9%, 2.68 [1.06–6.79], p=0.038) and CBD-dominant (41.1%, 3.10 [1.21–7.91], p=0.018) but not for THC+CBD (33.9% vs., 2.19 [0.85–5.64], p=0.103) vs. placebo (21.7%).

Only THC+CBD had superior sustained benefit versus placebo (Figures 2 and and3).3). THC+CBD was superior to placebo regarding 24-hour sustained pain freedom (28.0% vs. 10.7%, 3.45 [1.14–10.50], p=0.029), 24-hour sustained MBS freedom (46.0% vs. 25.0%, 2.83 [1.10–7.26], p=0.031), and 48-hour sustained MBS freedom (39.6% vs. 18.4%, 3.39 [1.24–9.32], p=0.018) but the difference for 48-hour sustained pain freedom was non-significant (22.9% vs 10.2%, 2.77 [0.84–9.12], p=0.094). THC-dominant and CBD-dominant were not different from placebo regarding 24- or 48-hour sustained pain freedom or sustained MBS freedom. Adverse events are reported in Table 2. Mean subjective highness on a 0–10 scale at 1 hour was greatest with THC-dominant at 3.5 (SD 3.0), followed by 2.4 (SD 2.5) with THC+CBD, 1.5 (SD 2.0) with CBD-dominant, and 0.6 (SD 1.20) with placebo. At 2 hours subjective highness was reduced to 2.4 (SD 2.8) with THC-dominant, 1.3 (SD 1.9) with THC+CBD, 0.9 (SD 1.6) with CBD-dominant, and 0.4 (SD 1.0) with placebo. THC-dominant had the greatest euphoria, cognitive impairment and subjective highness, followed by THC+CBD, then CBD-dominant, and least with placebo. Across all 4 treatments there were no serious adverse events (Table 2).

Table 2:

Adverse Events

A) Rates of queried adverse events
PlaceboCBDTHC+CBDTHC
1 Hour (N)N=60N=56N=56N=58
Sleepiness16 (26.7%)21 (37.5%)25 (44.6%)24 (41.4%)
Euphoria4(7%)5 (8.9%)16 (28.6%)21 (36.2%)
Cognitive Impairment4(7%)8 (14.3%)12 (21.4%)20 (34.5%)
Highness (0–10) Mean (SD)0.6 (1.2)1.5 (2.0)2.4 (2.5)3.5 (3.0)
2 Hours (N)N=58N=57N=58N=61
Sleepiness22 (37.9%)29 (50.9%)18 (31.0%)26 (42.6%)
Euphoria1 (1.7%)2 (3.5%)6 (10.3%)17 (27.9%)
Cognitive Impairment3 (5.2%)4 (7.0%)7 (12.1%)17 (26.2%)
Highness (0–10) Mean (SD)0.4 (1.0)0.9 (1.6)1.3(1.9)24(2.8)
B) Categorized free response adverse events
PlaceboCBDTHC+CBDTHC
Participants reporting any adverse events3/60 (5.0%)11/56 (19.6%)11/56 (19.6%)18/58 (31.0%)
Adverse events reported at 1 hour071213
 Sedation0230
 Slowness0104
 Paresthesias0012
 Throat irritation0320
 Increased appetite0211
 Impaired cognition0021
 Very high0001
 Impaired focus0022
 Dizziness0002
 Dry mouth0210
Adverse events reported by ≥1% of participants at 2 hours3/58 (5.2%)4/57 (7.0%)4/58 (6.9%)11/61 (18.0%)
 Sedation0322
 Impaired focus1001
 Slowness0003
 Dizziness0022
 Throat irritation0110
 Dry mouth2010
 Paresthesias0001
 Increased appetite0001
 Impaired cognition0011
 Very high0000

Regarding blinding, correct identification of treatment at 2 hours was 29.3% (17/58) with placebo, 10.5% (6/57) with CBD-dominant, 13.1% (8/61) with THC-dominant, and 20.7% (12/58) with THC+CBD. Participants selected “unknown” 24% of the time; 28% of the time with placebo, 26% with CBD-dominant, 22% with THC+ CBD, and 21% with THC-dominant. While 50% of treatments could have contained THC (THC-dominant or THC+CBD) and in this study 50.9% (119/234) treatments contained THC, participants correctly identified that a treatment contained THC (selecting “THC-dominant”, “THC+CBD”, or “Either THC-dominant or THC+CBD”) slightly less than expected by chance, 46.6% (27/58) of the time after THC+CBD treatment and 49.2% (30/61) after THC-dominant treatment. Meanwhile, 32% of participants selected a THC-containing answer after treatment with CBD-dominant and 14% after treatment with placebo.

Discussion

In this study, the first randomized, double-blind, placebo-controlled trial testing the efficacy of cannabinoids for acute migraine, 6% THC+11% CBD was superior to placebo for pain relief, pain freedom and MBS at 2 hours, as well as freedom from photophobia and phonophobia at 2 hours and 24-hour sustained pain freedom and sustained MBS freedom and 48-hour sustained MBS freedom.

THC potencies (6%) were lower than typical with cannabis available via American dispensaries and subjective highness (2.4 out of 10 at 1 hour) with THC+CBD was roughly half of that reported by research participants using cannabis ad lib33, bolstering evidence that higher potencies and titrating to highness are unnecessary for medicinal benefit. THC+CBD had less euphoria, subjective cognitive impairment, and subjective highness than THC-dominant. CBD is reported to be a non-competitive, negative allosteric modulator of the CB1 receptor, by which CBD may decrease the psychoactive effects of THC3436. The blinding data, as well as THC+CBD having more acute migraine benefit with less psychoactive effects as compared to THC-dominant, suggests that this study’s findings are not explainable by expectation effects or unmasking due to psychoactive side effects.

THC+CBD’s efficacy for reducing photophobia and phonophobia but not nausea in this study may be in part explainable by lower time 0 rates of nausea (59.9%) than photophobia (91.9%) and phonophobia (87.0%) in this study. That THC+CBD reduced photophobia and phonophobia but not nausea or vomiting in this study demonstrates that THC+CBD’s effects on MBS are not explainable by THC’s established antiemetic effects37 (Figure 3).

Strengths of the study include generalizability by enrolling patients with both episodic (72.5%) and chronic (27.5%) migraine to best reflect the larger patient population; historically most acute migraine studies have excluded patients with chronic migraine. Limitations of this study include that the study only examined THC and CBD; minor cannabinoids and terpenes were not studied. It also only studied single potencies of THC of CBD and a single THC:CBD ratio.

Typical of acute migraine RCTs, this study did not assess repeated administrations or regular, long-term treatments to assess possible risks (including rates of development of medication overuse headache and cannabis use disorder) or benefits (including possible migraine preventive effects)29,38. A small body of literature shows improvements on patient reported outcomes when using cannabis-based medicinal products on a regular, longer term or preventive basis20,39. More research is needed to evaluate repeated administrations and regular, long-term use of cannabinoids for migraine.

Conclusion

In this first randomized, double-blind, placebo-controlled trial testing the efficacy of cannabinoids for the acute treatment of migraine, vaporized 6% THC+11% CBD cannabis flower was superior to placebo for pain relief, pain freedom, and MBS freedom at 2 hours as well as 24-hour sustained pain freedom and sustained MBS freedom and 48-hour sustained MBS freedom. Future research should include multicenter studies and long-term studies of benefits and risks with repeated use.

Acknowledgements

This study was sponsored by the Migraine Research Foundation, now administered by Hartford HealthCare. The project described was partially supported by the UCSD Academic Senate and the National Institutes of Health, Grant UL1TR001442. The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH. Study drug was provided by the National Institute on Drug Abuse Drug Supply Program. The authors thank the National Institute on Drug Abuse Drug Supply Program and the National Center for Natural Products Research at the University of Mississippi School of Pharmacy. Vaporizers were donated by Storz & Bickel GmbH & Co. KG, which did not provide any financial support of this study and was not involved in the design or conduct of the study or manuscript preparation. Thanks to Dr. Brooke Towne, MD, Phirum Nguyen, Gayle Dizon, Jay Patel, Brett Taylor, and Megan Sweeney for assistance with conducting this study. Thank you to all the participants who volunteered for this study.

Disclosures:

NMS has received research funding from Migraine Research Foundation, Novaremed, UCSD Academic Senate, UCSD Department of Anesthesiology RAG, and NIH CTSA Grant UL1TR000100 and has consulted for Eli Lilly & Co., Averitas, Syneos, Schedule 1 Therapeutics, Vectura Fertin, and ShiraTronics. He serves on the editorial board of Pain Medicine (Oxford University Press). DCB has been a consultant for Abbvie, Amgen, Biohaven, Lilly, Lundbeck, Teva, Theranica. She has received grant support from the US Food and Drug Association and National Headache Foundation. She is an editor for Current Pain and Headache Reports. MSW, TM, EL, LL and MS have no disclosures to report.

Footnotes

Trial Registration Number: NCT04360044

References

1. Safiri S, Pourfathi H, Eagan A, et al. Global, regional, and national burden of migraine in 204 countries and territories, 1990 to 2019. Pain. 2022;163(2):E293–E309. doi: 10.1097/J.PAIN.0000000000002275 [PubMed] [CrossRef] [Google Scholar]

2. Ailani J, Burch RC, Matthew |, Robbins S. The American Headache Society Consensus Statement: Update on integrating new migraine treatments into clinical practice. Headache. 2021;61:1021–1039. doi: 10.1111/head.14153 [PubMed] [CrossRef] [Google Scholar]

3. Serrano D, Buse DC, Manack Adams A, Reed ML, Lipton RB. Acute treatment optimization in episodic and chronic migraine: results of the American Migraine Prevalence and Prevention (AMPP) Study. Headache. 2015;55(4):502–518. doi: 10.1111/HEAD.12553 [PubMed] [CrossRef] [Google Scholar]

4. Holland S, Fanning KM, Serrano D, Buse DC, Reed ML, Lipton RB. Rates and reasons for discontinuation of triptans and opioids in episodic migraine: Results from the American Migraine Prevalence and Prevention (AMPP) study. J Neurol Sci. 2013;326(1–2):10–17. doi: 10.1016/J.JNS.2012.12.020 [PubMed] [CrossRef] [Google Scholar]

5. Sexton M, Cuttler C, Finnell JS, Mischley LK. A Cross-Sectional Survey of Medical Cannabis Users: Patterns of Use and Perceived Efficacy. Cannabis Cannabinoid Res. 2016;1(1):131–138. doi: 10.1089/can.2016.0007 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

6. Gorelick DA. Cannabis-Related Disorders and Toxic Effects. Hardin CC, ed. N Engl J Med. 2023;389(24):2267–2275. doi: 10.1056/NEJMra2212152 [PubMed] [CrossRef] [Google Scholar]

7. Poudel S, Quinonez J, Choudhari J, et al. Medical Cannabis, Headaches, and Migraines: A Review of the Current Literature. Cureus. 2021;13(8). doi: 10.7759/CUREUS.17407 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

8. Akerman S, Kaube H, Goadsby PJ. Anandamide is able to inhibit trigeminal neurons using an in vivo model of trigeminovascular-mediated nociception. J Pharmacol Exp Ther. 2004;309(1):56–63. doi: 10.1124/jpet.103.059808\rjpet.103.059808 [pii] [PubMed] [CrossRef] [Google Scholar]

9. Akerman S, Holland PR, Goadsby PJ. Cannabinoid (CB1) receptor activation inhibits trigeminovascular neurons. J Pharmacol Exp Ther. 2007;320(1):64–71. doi: 10.1124/jpet.106.106971 [PubMed] [CrossRef] [Google Scholar]

10. Baron EP. Comprehensive review of medicinal marijuana, cannabinoids, and therapeutic implications in medicine and headache: What a long strange trip it’s beenHeadache. 2015;55(6):885–916. doi: 10.1111/head.12570 [PubMed] [CrossRef] [Google Scholar]

11. 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):1139–1186. doi: 10.1111/head.13345 [PubMed] [CrossRef] [Google Scholar]

12. McGeeney BE. Cannabinoids and hallucinogens for headache. Headache. 2013;53(3):447–458. doi: 10.1111/head.12025 [PubMed] [CrossRef] [Google Scholar]

13. Akerman S, Holland PR, Lasalandra MP, Goadsby PJ. Endocannabinoids in the Brainstem Modulate Dural Trigeminovascular Nociceptive Traffic via CB1 and “Triptan” Receptors: Implications in Migraine. J Neurosci. 2013;33(37):14869–14877. doi: 10.1523/JNEUROSCI.0943-13.2013 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

14. Kazemi H, Rahgozar M, Speckmann E-J, Gorji A. Effect of cannabinoid receptor activation on spreading depression. Iran J Basic Med Sci. 2012;15(4):926–936. http://www.ncbi.nlm.nih.gov/pubmed/23493641%5Cnhttp://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=PMC3586901. [PMC free article] [PubMed] [Google Scholar]

15. Aviram J, Vysotski Y, Berman P, Lewitus GM, Eisenberg E, Meiri D. Migraine Frequency Decrease Following Prolonged Medical Cannabis Treatment: A Cross-Sectional Study. Brain Sci. 2020;10(6):1–15. doi: 10.3390/BRAINSCI10060360 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

16. Stith S, Diviant J, Brockelman F, et al. Alleviative effects of Cannabis flower on migraine and headache. J Integr Med. 2020;18(5):416–424. doi: 10.1016/J.JOIM.2020.07.004 [PubMed] [CrossRef] [Google Scholar]

17. Baraldi C, Lo Castro F, Negro A, et al. Oral Cannabinoid Preparations for the Treatment of Chronic Migraine: A Retrospective Study. Pain Med. 2022;23(2):396–402. doi: 10.1093/pm/pnab245 [PubMed] [CrossRef] [Google Scholar]

18. Rhyne DN, Anderson SL, Gedde M, Borgelt LM. Effects of Medical Marijuana on Migraine Headache Frequency in an Adult Population. Pharmacotherapy. 2016;36(5):505–510. doi: 10.1002/phar.1673 [PubMed] [CrossRef] [Google Scholar]

19. Cuttler C, Spradlin A, Cleveland MJ, Craft RM. Short- and Long-Term Effects of Cannabis on Headache and Migraine. J pain. 2020;21(5–6):722–730. doi: 10.1016/j.jpain.2019.11.001 [PubMed] [CrossRef] [Google Scholar]

20. Nicholas M, Erridge S, Bapir L, et al. UK medical cannabis registry: assessment of clinical outcomes in patients with headache disorders. Expert Rev Neurother. 2023;23(1):85–96. doi: 10.1080/14737175.2023.2174017 [PubMed] [CrossRef] [Google Scholar]

21. Lochte BC, Beletsky A, Samuel NK, Grant I. The Use of Cannabis for Headache Disorders. Cannabis Cannabinoid Res. 2017;2(1):61–71. doi: 10.1089/can.2016.0033 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

22. Cady RK, McAllister PJ, Spierings EL, et al. A Randomized, Double-Blind, Placebo-Controlled Study of Breath Powered Nasal Delivery of Sumatriptan Powder (AVP-825) in the Treatment of Acute Migraine (The TARGET Study). Headache. 2015;55(1):88–100. doi: 10.1111/head.12472 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

23. 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):785–796. [PubMed] [Google Scholar]

24. Wallace MS, Marcotte TD, Umlauf A, Gouaux B, Atkinson JH. Efficacy of Inhaled Cannabis on Painful Diabetic Neuropathy. J Pain. 2015;16(7):616–627. doi: 10.1016/j.jpain.2015.03.008 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

25. Wallace MS, Marcotte TD, Atkinson JH, Padovano HT, Bonn-Miller M. A Secondary Analysis from a Randomized Trial on the Effect of Plasma Tetrahydrocannabinol Levels on Pain Reduction in Painful Diabetic Peripheral Neuropathy. J pain. 2020;21(11–12):1175–1186. doi: 10.1016/j.jpain.2020.03.003 [PubMed] [CrossRef] [Google Scholar]

26. Headache Classification Committee of the International Headache Society. The International Classification of Headache Disorders, 3rd edition. Cephalalgia. 2018. doi: 10.1177/0333102417738202 [PubMed] [CrossRef] [Google Scholar]

27. Obeid JS, McGraw CA, Minor BL, et al. Procurement of shared data instruments for Research Electronic Data Capture (REDCap). J Biomed Inform. 2013;46(2):259–265. doi: 10.1016/J.JBI.2012.10.006 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

28. Foltin RW, Brady J V., Fischman MW. Behavioral analysis of marijuana effects on food intake in humans. Pharmacol Biochem Behav. 1986;25(3):577–582. doi: 10.1016/0091-3057(86)90144-9 [PubMed] [CrossRef] [Google Scholar]

29. Diener HC, Tassorelli C, Dodick DW, et al. Guidelines of the International Headache Society for controlled trials of acute treatment of migraine attacks in adults: Fourth edition. Cephalalgia. 2019;39(6):687–710. doi: 10.1177/0333102419828967 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

30. Ware JH, Harrington D, Hunter DJ, D’Agostino RB. Missing Data. N Engl J Med. 2012;367(14):1353–1354. doi: 10.1056/NEJMsm1210043 [CrossRef] [Google Scholar]

31. Thabane L, Mbuagbaw L, Zhang S, et al. A tutorial on sensitivity analyses in clinical trials: the what, why, when and how. BMC Med Res Methodol. 2013;13:92. doi: 10.1186/1471-2288-13-92 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

32. Cro S, Morris TP, Kenward MG, Carpenter JR. Sensitivity analysis for clinical trials with missing continuous outcome data using controlled multiple imputation: A practical guide. Stat Med. 2020;39(21):2815–2842. doi: 10.1002/sim.8569 [PubMed] [CrossRef] [Google Scholar]

33. Marcotte TD, Umlauf A, Grelotti DJ, et al. Driving Performance and Cannabis Users’ Perception of Safety: A Randomized Clinical Trial. JAMA psychiatry. 2022;79(3):201–209. doi: 10.1001/jamapsychiatry.2021.4037 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

34. Laprairie RB, Bagher AM, Kelly MEM, Denovan-Wright EM. Cannabidiol is a negative allosteric modulator of the cannabinoid CB1 receptor. Br J Pharmacol. 2015;172(20):4790–4805. doi: 10.1111/bph.13250 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

35. Pertwee RG, Ross RA, Craib SJ, Thomas A. (−)-Cannabidiol antagonizes cannabinoid receptor agonists and noradrenaline in the mouse vas deferens. Eur J Pharmacol. 2002;456(1–3):99–106. doi: 10.1016/s0014-2999(02)02624-9 [PubMed] [CrossRef] [Google Scholar]

36. Schubart CD, Sommer IEC, van Gastel WA, Goetgebuer RL, Kahn RS, Boks MPM. Cannabis with high cannabidiol content is associated with fewer psychotic experiences. Schizophr Res. 2011;130(1–3):216–221. doi: 10.1016/j.schres.2011.04.017 [PubMed] [CrossRef] [Google Scholar]

37. Sallan SE, Zinberg NE, Frei E. Antiemetic Effect of Delta-9-Tetrahydrocannabinol in Patients Receiving Cancer Chemotherapy. N Engl J Med. 1975;293(16):795–797. doi: 10.1056/NEJM197510162931603 [PubMed] [CrossRef] [Google Scholar]

38. U.S. Department of Health and Human Services: Food and Drug Administration. Migraine: Developing Drugs for Acute Treatment- Guidance for Industry.; 2018. https://www.fda.gov/downloads/drugs/guidances/ucm419465.pdf. [Google Scholar]

39. Okusanya BO, Lott BE, Ehiri J, McClelland J, Rosales C. Medical Cannabis for the Treatment of Migraine in Adults: A Review of the Evidence. Front Neurol. 2022;13:871187. doi: 10.3389/FNEUR.2022.871187/FULL [PMC free article] [PubMed] [CrossRef] [Google Scholar]

Articles from medRxiv are provided here courtesy of Cold Spring Harbor Laboratory Preprints

Vaporized Cannabis versus Placebo for Acute Migraine: A Randomized Controlled Trial (2024)

FAQs

How effective is inhaled cannabis for acute migraine treatment? ›

Ninety-two participants were enrolled and randomized, and 247 migraine attacks were treated. THC+CBD was superior to placebo at achieving pain relief (67.2% vs 46.6%, Odds Ratio [95% Confidence Interval] 2.85 [1.22, 6.65], p=0.016), pain freedom (34.5% vs. 15.5%, 3.30 [1.24, 8.80], p=0.017) and MBS freedom (60.3% vs.

Which FDA approved both to treat acute migraine attacks and as preventive therapy? ›

Rimegepant was the first oral treatment option of its class to treat acute migraine attacks, and now, this CGRP small molecule antagonist is now indicated to prevent migraine attacks as well.

Does CBG help with migraines? ›

Some research suggests that CBD may help treat migraine headaches. A growing body of research suggests that CBD may help relieve pain, particularly neurological pain, linked with various conditions. Specifically, CBD oil has promise as a treatment for migraine, as the American Migraine Foundation report.

What migraine device is FDA cleared? ›

The U.S. Food and Drug Administration (FDA) recently cleared a new neuromodulation device, called HeadaTerm 2, for migraine prevention, and it'll be available soon without a prescription. Migraine is a common and disabling disease that affects more than 40 million people in the United States.

What type of cannabis is best for migraines? ›

In the ID Migraine™ questionnaire, hybrid strains of cannabis, of which "OG Shark," a high THC/THCA, low CBD/CBDA, and strains with predominant terpenes β-caryophyllene and β-myrcene, were most preferred in the headache and migraine groups [20].

What is the inhalable medicine for migraines? ›

Zolmitriptan nasal spray is used to treat the symptoms of migraine headaches (severe, throbbing headaches that sometimes are accompanied by other symptoms such as nausea and sensitivity to sound and light). Zolmitriptan is in a class of medications called selective serotonin receptor agonists.

What is the new treatment for acute migraines? ›

Ubrogepant received US Food and Drug Administration (FDA) approval for the treatment of acute migraine in adults in December 2019 [108]. Rimegepant – Rimegepant is given at 75 mg as a single oral dose. Clinical trial evidence supports the efficacy of rimegepant for acute migraine treatment.

Why was Aimovig discontinued? ›

AIMOVIG was discontinued in most reported cases of constipation with serious complications.

What is the #1 migraine medication? ›

UBRELVY QUICKLY ELIMINATES MIGRAINE PAIN

UBRELVY is the #1 prescribed branded treatment for migraine attacks in adults—it's not for prevention of migraine.

Is sativa or indica better for migraines? ›

Sativa is one of the most popular medical cannabis strains for cluster headaches, migraines, injuries and other types of head pains. It contains moderate amounts of THC and CBD, whose potential effects are believed to help with chronic pain management.

What terpenes are good for migraines? ›

β-caryophyllene (BCP), the most dominant terpene found in cannabis strains found to be most effective in treating migraines, is a potent CB2 agonist. Research supports the use of BCP in various chronic pain conditions.

What is a thunderclap migraine? ›

Overview. Thunderclap headaches live up to their name, striking suddenly like a clap of thunder. The pain of these severe headaches peaks within 60 seconds. Thunderclap headaches are uncommon, but they can warn of potentially life-threatening conditions — usually having to do with bleeding in and around the brain.

What is the new migraine drug NICE? ›

Atogepant is a new type of anti-calcitonin gene-related peptide (CGRP) drug, which is specifically designed to treat migraines. They work by blocking the receptor of the CGRP protein.

What is the Israeli device for migraines? ›

Reddy's Laboratories has forayed into the digital therapeutics market of Europe by unveiling Israeli firm Theranica's drug-free, non-invasive migraine management wearable device Nerivio in Germany through a step-down subsidiary. The Hyderabad-based drugmaker had introduced the device last year in India.

What is the new migraine medicine called? ›

The newest oral drugs for the acute treatment of migraine are Nurtec ODT (rimegepant) and Ubrelvy (ubrogepant), both orally-administered calcitonin gene-related peptide (CGRP) receptor antagonists (gepants).

What is the duration of action of inhaled cannabis? ›

You will feel the effects from seconds to a few minutes of inhaling. Full effects can peak within 30 minutes. Effects can last up to 6 hours after use.

How long does it take for inhaled cannabis to work? ›

Inhalation (smoking or vaping)

When cannabis is inhaled into the lungs it passes directly into the bloodstream and then the brain. The effects are usually quickly felt by the brain and body, from seconds to within minutes of inhaling. The full effects from one inhalation can take up to 10 to 30 minutes to peak.

How much CBD does it take to get rid of a migraine? ›

Usually, this means taking about 50 – 100mg of CBD oil per day. I have found this to be a good dose when trying to reduce the frequency of migraine attacks. When having a migraine attack, I encourage the use of inhaled CBD as opposed to CBD oil.

What is the intranasal opioid spray for migraines? ›

Butorphanol nasal spray is used to relieve pain severe enough to require opioid treatment and when other pain medicines did not work well enough or cannot be tolerated. It belongs to the group of medicines called narcotic analgesics (pain medicines). Butorphanol acts on the central nervous system (CNS) to relieve pain.

Top Articles
Latest Posts
Article information

Author: Sen. Ignacio Ratke

Last Updated:

Views: 6192

Rating: 4.6 / 5 (56 voted)

Reviews: 87% of readers found this page helpful

Author information

Name: Sen. Ignacio Ratke

Birthday: 1999-05-27

Address: Apt. 171 8116 Bailey Via, Roberthaven, GA 58289

Phone: +2585395768220

Job: Lead Liaison

Hobby: Lockpicking, LARPing, Lego building, Lapidary, Macrame, Book restoration, Bodybuilding

Introduction: My name is Sen. Ignacio Ratke, I am a adventurous, zealous, outstanding, agreeable, precious, excited, gifted person who loves writing and wants to share my knowledge and understanding with you.