Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 Jan 1.
Published in final edited form as: JAMA Psychiatry. 2023 Jan 1;80(1):84–92. doi: 10.1001/jamapsychiatry.2022.4020

Clinical Trial Design Challenges and Opportunities for Emerging Treatments for Opioid Use Disorder A Review

Brian D Kiluk 1, Bethea A Kleykamp 1, Sandra D Comer 1, Roland R Griffiths 1, Andrew S Huhn 1, Matthew W Johnson 1, Kyle M Kampman 1, Marco Pravetoni 1, Kenzie L Preston 1, Ryan Vandrey 1, Cecilia L Bergeria 1, Michael P Bogenschutz 1, Randall T Brown 1, Kelly E Dunn 1, Robert H Dworkin 1, Patrick H Finan 1, Peter S Hendricks 1, Elisabeth J Houtsmuller 1, Thomas R Kosten 1, Dustin C Lee 1, Frances R Levin 1, Aimee McRae-Clark 1, Charles L Raison 1, Kurt Rasmussen 1, Dennis C Turk 1, Roger D Weiss 1, Eric C Strain 1
PMCID: PMC10297827  NIHMSID: NIHMS1907289  PMID: 36449315

Abstract

IMPORTANCE

Novel treatments for opioid use disorder (OUD) are needed to address both the ongoing opioid epidemic and long-standing barriers to existing OUD treatments that target the endogenous μ-opioid receptor (MOR) system. The goal of this review is to highlight unique clinical trial design considerations for the study of emerging treatments for OUD that address targets beyond the MOR system. In November 2019, the Analgesic, Anesthetic, and Addiction Clinical Trial Translations, Innovations, Opportunities, and Networks (ACTTION) public-private partnership with the US Food and Drug Administration sponsored a meeting to discuss the current evidence regarding potential treatments for OUD, including cannabinoids, psychedelics, sedative-hypnotics, and immunotherapeutics, such as vaccines.

OBSERVATIONS

Consensus recommendations are presented regarding the most critical elements of trial design for the evaluation of novel OUD treatments, such as: (1) stage of treatment that will be targeted (eg, seeking treatment, early abstinence/detoxification, long-term recovery); (2) role of treatment (adjunctive with or independent of existing OUD treatments); (3) primary outcomes informed by patient preferences that assess opioid use (including changes in patterns of use), treatment retention, and/or global functioning and quality of life; and (4) adverse events, including the potential for opioid-related relapse or overdose, especially if the patient is not simultaneously taking maintenance MOR agonist or antagonist medications.

CONCLUSIONS AND RELEVANCE

Applying the recommendations provided here as well as considering input from people with lived experience in the design phase will accelerate the development, translation, and uptake of effective and safe therapeutics for individuals struggling with OUD.


Opioid use disorder (OUD) is a major cause of disease burden, leading to increased pregnancy or birth complications, viral infections, and fatal overdoses.13 The 3 effective and safe medications for treating OUD (MOUD) act through the μ-opioid receptor (MOR), the primary target for opioids misused for their rewarding effects.4 The MOR agonists methadone or buprenorphine and the MOR antagonist naltrexone are the standard of care for OUD because they reduce risk of relapse, overdose deaths, infections, and criminal behavior,5 but discontinuation and relapse still exceed 50% within 6 months.68 Furthermore, each of these MOUDs have different induction and dosing procedures as well as regulatory, policy, and patient-level barriers that have hindered patient access and retention.9 Thus, OUD treatment options need expansion through development of novel stand-alone therapies or adjuncts to existing MOR-based MOUDs.1012

A critical step in developing novel treatments for OUD is the completion of randomized clinical trials (RCTs). However, the inherent features of OUD, including a pronounced physical dependence and a high risk of overdose, suggest the design of these trials will likely need to differ from designs used to evaluate existing treatments for OUD. There is not a strong consensus in the OUD field concerning standardized key trial design decisions or outcome measures. Given the importance of this topic and the need for new and novel OUD treatments, a meeting sponsored by the Analgesic, Anesthetic, and Addiction ClinicalTrialTranslations, Innovations, Opportunities, and Networks (ACTTION) public-private partnership with the US Food and Drug Administration (FDA) was convened in November 2019 to discuss study design considerations unique to 4 candidate medication categories for OUD that do not directly target the MOR system: cannabinoids, psychedelics, sedative-hypnotics, and immunotherapeutics;a summary of highlights from the meeting has been previously published.13 This article reviews the key trial considerations derived from that meeting and provides consensus considerations and recommendations for studies of non-MOR–based treatments for OUD.

Methods

The ACTTION Consortium for Addiction Research on Efficacy and Safety (CARES) meeting included participants from academia, government, and nonprofit organizations selected on the basis of their research, clinical, or administrative expertise relevant to the candidate medication categories or clinical trials of OUD treatments. There was no direct participation from any pharmaceutical company. Meeting details, including agenda, goals, list of attendees, presentations, and transcripts of discussion, are available on the CARES website.14 The following considerations and recommendations were informed by the meeting presentations and discussions, literature reviews, and coauthors’ feedback on iterative revisions of drafts of this article.

Discussion

Study Planning

Study planning should begin by specifying the stage in OUD treatment targeted by the intervention(s), as this decision will influence all subsequent design decisions. The core stages in the OUD treatment and recovery trajectory can be conceptualized as (1) current active use of opioids; (2) acute abstinence, nonmedically supervised withdrawal, and/or supervised medical withdrawal; (3) early recovery (eg, less than 6 months of abstinence with or without opioid agonist or antagonist treatment); and (4) sustained recovery (eg, abstinence from illicit opioid use for at least 6 months). Each stage has unique treatment needs, and study planning should consider whether the novel treatment will be adjunctive to existing regulatory agency-approved MOUD, which may be essential for those with physical dependence and withdrawal symptoms. The need for adjunctive treatment, including harm reduction strategies, such as naloxone training to prevent fatal overdose, would be essential for clinically unstable patients.

Historically, few OUD trials have incorporated the preferences of patients, and per patient-focused drug development,15 we recommend using input from people with lived experience to guide the choice of primary and secondary outcomes. For instance, although treatment retention was found as the most reported outcome across 60 OUD trials, many patients report an eagerness to complete therapy and end agonist treatment as a main goal.16 Strategies for incorporating patient perspectives into study planning include focus groups, interviews, online surveys, workshops, social media listening, and community-based participatory research strategies.17,18 Guidance on methods for engaging patients and other relevant stakeholders are described elsewhere.19

Study Design

Intervention (Including Randomization, Blinding, and Dosing)

Trial designs will be dictated to a large extent by the stage of treatment that the intervention is targeting as well as the unique properties of the intervention under evaluation. The Table gives an overview of specific considerations for the 4 types of emerging medication treatments reviewed here.2054 The National Institute on Drug Abuse has identified additional emerging areas of interest for OUD treatment development that target a range of novel pharmacological mechanisms of action, such as respiratory stimulants, γ-aminobutyric acid metabotropic receptor family B agonists, and ghrelin antagonists.11 Discussing all emerging treatments, including nonmedication interventions (eg, repetitive transcranial magneticstimulation55), was beyond the scope of this meeting, yet many of the considerations and recommendations described here also apply to these other approaches. Each of these emerging treatments has specific characteristics that influence study design choices, including dosing, mode of administration, and timing of intervention relative to treatment stage.

Table.

Unique Considerations Associated With 4 Emerging Treatments for Opioid Use Disorder

Consideration Cannabis and cannabinoids Psychedelics Sedative-hypnotics Immunotherapeutics (vaccines and monoclonal antibodies)
Rationale The endocannabinoid and opioid systems interact with some subtypes of cannabinoid receptors that influence the rewarding effects of opioids. Some uncontrolled observational research has suggested that cannabis and cannabinoids can have a substitution effect on opioid use behavior.2027 Classic psychedelics (serotonin 2A receptor agonists) have been associated with reduced substance use in naturalistic and clinical settings, with the strongest evidence for LSD as a treatment of alcoholism. Candidate psychological mechanisms of action include awe, cognitive flexibility, and insight; candidate biological mechanisms include inflammation and brain network functioning.2834 Sleep is a basic biological system that can be affected by opioid use and can also affect the trajectory of opioid use. Sleep dysfunction is a common issue across all substance use disorders. Sleep disturbance can have profound effects on a patient’s life, including ability to cope with craving, and can affect the cognitive effort associated with opioid abstinence.3541 Active (vaccination) and passive (transfer of premade antibodies) immunization strategies rely on the presence of drug-specific antibodies to selectively bind to target opioids in plasma and prevent drugs from crossing the blood-brain barrier and reaching the brain. By reducing the concentration of free (unbound) opioids in the brain, vaccines and monoclonal antibodies reduce opioids’ pharmacological effects.4250
Types Cannabis is a complex chemical entity that contains >100 botanically derived phytocannabinoids, each of which can be synthesized or isolated. LSD, psilocybin (found in Psilocybe mushrooms), mescaline (found in peyote and other cacti), and dimethyltryptamine (found in ayahuasca). Orexin-1 or 1/2 antagonists; tricyclics (Doxepin); antipsychotics (Quetiapine); melatonin; mirtazapine; or ramelteon. Benzodiazepines or benzodiazepinelike drugs (eg, Zolpidem) are often not used due to risk for misuse. Individual and multivalent vaccines targeting specific types of opioids, including oxycodone, heroin, and fentanyl. Individual and multivalent monoclonal antibodies formulations against various opioids.
Example human trials (patients with OUD) 60 Participants with OUD were randomized to receive dronabinol, 30 mg/d, or placebo during inpatient supervised withdrawal followed by oral naltrexone induction and extended-release maintenance for 5 wk. Dronabinol reduced the severity of opioid withdrawal during acute withdrawal but had no effect on rates of extended-release naltrexone treatment induction and retention.24 74 Male parolees with a history of chronic heroin use participated in a 4- to 6-wk residential program involving preparatory therapy in conjunction with a single high-dose administration of LSD (300–450 μg) compared with treatment as usual (ie, outpatient weekly group therapy). The LSD treatment was well tolerated, and biologically verified continuous abstinence was significantly greater in the LSD condition than control condition.51 137 Participants receiving methadone for OUD who reported a PSQI score of ≥6 were randomized to trazodone, 50 mg, or placebo. They completed 2 night home polysomnography sessions that were separated by 1 mo. Trazadone did not significantly affect subjective sleep ratings or most objective sleep outcomes.52 347 Participants in Iran using morphine were injected with 3 doses of a morphine vaccine at baseline, 30 d, and 60 d. The concentration of antimorphine antibodies was correlated with number of morphine injections detected at 30 d, reaching their peak by 3 mo after first injections and did not return to baseline levels by 1 y.53 An ongoing clinical trial of an oxycodone vaccine is currently being conducted in the US.54
Key challenges Choice of study drug, including mode and route of administration, which is affected by product variation across states/countries; access to study drug and phase II or higher trials challenging because of schedule I status; clarity is needed around objective of intervention. Preparation and therapeutic alliance between study participant and researchers is necessary to reduce psychological risks; active placebo might not be feasible and could be replaced using subtherapeutic doses of tested compound or an active control; access to study drug and phase II or higher trials challenging because of schedule I status. Choice of study setting (and associated sleep technology) affects the quality of sleep measures; rapidly changing technology with different levels of comfort for participants; the effect on opioid use will depend on presence of acute or chronic sleep agent dosing as well as the treatment stage in which it is tested. Potential for compensatory use of opioids; design vaccines and monoclonal antibodies that do not interfere with opioid agonists or antagonists or other critical medications; equally reduced rewarding effects of opioids as well as respiratory depressant effects.
Potential exclusion criteria Current or previous dependence on or tolerance to cannabis or presence of alcohol or other substance use disorders or comorbid psychiatric disorders (eg, psychosis, anxiety). Uncontrolled hypertension or comorbid psychiatric disorders (eg, bipolar or psychotic disorders); patients taking medications with perceptual effects or effects on serotonergic function. Existing sleep difficulties or diagnosed sleep disorders that are not the primary target of the intervention (eg, severe central or obstructive sleep apnea) as well as alcohol or other substance use disorders; active use of illicit opioids as couse with some sedative-hypnotics may be life threatening. Polyopioid use may require use of multivalent vaccines against multiple drug targets (eg, heroin/fentanyl). Potential need for using diagnostic assays or predictive biomarkers to select patients most suitable for vaccine treatment for comorbid immune-compromising diseases.
Outcomes and measures of interest unique to the intervention Misuse risk, including subjective ratings of liking or wanting cannabis, cognitive performance. Acute mystical/transcendent/awe experiences, meaningful/insightful nature of experience, life functioning, quality of life, affect, prosocial behavior, persisting adverse effects, including perceptual disturbance, cardiovascular outcomes (eg, heart rate, blood pressure), headaches, psychological distress, positive and negative effects on cooccurring psychiatric disorders, risk for misuse, including use outside of controlled research settings. Affect, opioid craving, objective and/or subjective measurement of sleep quality and sleep architecture measured using polysomnography, ambulatory monitors (eg, Sleep Profiler, which can be used at home and yield polysomnography-level outcomes), actigraphy (can be used as a primary or secondary measure of sleep by tracking movement, room lighting, etc.). Other wearable technology (eg, Oura Ring; Watchpat), ecological momentary assessment, consensus sleep diary, and retrospective questionnaires (eg, PSQI or Insomnia Severity index). Immunological and pharmacokinetic outcomes, including antibody response and the effect of antibodies on distribution of drug in plasma (vaccine efficacy in reducing the effects of clinically relevant doses of target drugs depends on the quantity and quality of the antibody response). Other measures include effect of vaccine or monoclonal antibodies on drug-liking behaviors.

Abbreviations: LSD, lysergic acid diethylamide; OUD, opioid use disorder; PSQI, Pittsburgh Sleep Quality Index.

For drug development, the criterion-standard efficacy and safety studies are double-blind, placebo-controlled RCT designs. However, for OUD, these designs face ethical concerns of a placeboonly condition and challenges in blinding treatment groups. Additional research designs that could be considered include adaptive or pragmatic trials and the use of real-world data as primary or secondary outcomes.56,57 Regardless of the specifics of blinding and ran-domization, we recommend that efforts to examine novel compounds be paired with some form of standardized and efficacious psychosocial support, including in-person or digital treatment modules, to mitigate the risk that patients are left with no treatment if a compound fails.58

Comparators

The severe nature of the opioid physical dependence syndrome means that a placebo-controlled trial in the absence of an agonist MOUD might be unsafe or unfeasible for patients who are in early abstinence and at risk of opioid withdrawal symptoms, relapse, or overdose. Relevant alternative types of comparators include (1) low or subtherapeutic doses of study medication, (2) ascending doses of study medication, (3) standard-of-care pharmacologic or nonpharmacologic treatments in a comparative effectiveness trial design, or (4) a combination of different comparators.

The type of comparator will also influence whether the objective of the clinical trial is to test superiority or noninferiority between different treatment conditions. Investigators may choose to provide an MOUD as a plat form therapy for all participants while comparing an active vs placebo adjunctive medication using a superiority trial design (eg, a sleep agent compared with placebo for those stabilized with methadone). Ethical concerns related to place bodosing could also increase the appeal of noninferiority trials, although these are more complex in design and analysis than superiority trials, with challenges described elsewhere.59

Study Setting

RCTs of MOR-based MOUDs have been traditionally completed on an outpatient basis in settings, such as opioid treatment programs or medical offices, because of inherent restrictions on MOUD prescribing and dispensing. Some emerging treatments, such as sedative-hypnotics or vaccines, may have fewer regulatory or medical requirements compared with MOR-based treatments and therefore may afford more flexibility in the study designs and open opportunities for novel approaches.60,61 Methods for remote data collection have advanced considerably during the SARS-CoV-2 pandemic, expanding possible approaches to collecting substance use outcomes (eg, remotely collected breathalyzer data for alcohol or tobacco use).6264 Recent parallel efforts to leverage nonspecialized care professionals to expand the OUD treatment infrastructure, including health care professionals,65,66 may further bolster innovation. However, these approaches may not be useful in all cases; the study of some agents may require even more intensive in-person designs compared with traditional OUD clinical trials. The in-person interactions and monitoring required for safe delivery and evaluation of some novel treatments present challenges to conducting clinical trials on a larger scale, an issue the field has acknowledged and begun to address with more scalable intervention paradigms.67,68

Participant Characteristics

Participant selection in the form of inclusion and exclusion criteria are essential for ensuring that a trial targets the population of interest, minimizes variance in outcomes because of factors other than the intervention, and supports future meta-analyses. At minimum, we recommend that the following categories be addressed in the study inclusion and exclusion criteria and/or baseline data collection associated with the study: (1) opioid use variables, including historical (lifetime) and current (past year) opioid use behavior, including type, timing, amount, and route of administration of opioid(s), previous experience with opioid overdose, including hospitalization, OUD treatment history, and degree of OUD severity; (2) historical or current alcohol and other substance use disorders, including prior use of target medication; (3) medical history, including prescribed medications in past 90 days and concomitant medical and psychiatric conditions; and (4) psychosocial variables (eg, problems resulting from opioid use, including incarceration). In addition, basic patient demographic characteristics (eg, age, sex, gender, race and ethnicity, and socioeconomic status) should be collected with awareness of specific populations that are at risk of developing OUD or those who experience disparate consequences, including individuals with mental health disorders,69 youth and young adults,70 military veterans,71 pregnant women,72 racial and ethnic minority populations,73,74 and individuals from particular geographic regions (eg, US Appalachian and Southern states).75,76 Limitations should be considered when selecting eligibility criteria depending on specific safety considerations associated with the intervention under study.

Outcome Measures

The type of efficacy outcomes chosen for a trial depends on the goal of the trial (eg, targeted phase of OUD treatment, key comparators). Literature reviews have noted that primary and secondary outcomes and their associated measures vary widely across clinical trials for OUD.16,77 Opioid abstinence and treatment retention have been the most common primary end points in clinical trials for OUD and other substance use disorders.78 However, there is an evolving understanding of the importance of continuous measures of opioiduse, including changes in use patterns, such as the frequency, duration, and amount of use.

The degree to which these different, but important, outcomes are clinically meaningful is still being debated.1,16,79,80 Currently there are no criterion-standard outcomes in OUD trials. Thus, the below recommendations are meant to functionas guide posts when choosing outcomes.

Primary Outcomes

The dichotomous outcome of opioid abstinence, defined as no detected or self-reported use within an assessment window, has been the most common measure of opioid use behavior in clinical trials.78 According to the FDA Guidance for Industry regarding end points for demonstrating effectiveness of drugs for treatment of OUD,78 drug use patterns other than abstinence can be used as thresholds to define treatment response. Measurement of such response-defining thresholds must be specified, and evidence from clinical trials, longitudinal observation studies, or other sources are needed to support the clinical benefit of a given drug use pattern (ie, reduction).78 We recommend that both abstinence and patterns of opioid use be measured and that clear responder criteria be specified for each, with the potential for a grace period. For trials that identify opioid abstinence as the primary outcome, we recommend opioid use be assessed using objective (eg, urinalysis) and subjective (eg, patient, clinician, and/or observer) measures.77 The field is currently moving to less frequent objective testing of these outcomes for practical reasons and to reduce the burden on participants. We recommend that decisions regarding frequency of testing be based on the clinical stability of the patient population, the pharmacological properties of treatment, and participation burden.

Trials of MOR-based treatments demonstrate that retention in treatment longer than 6 months is associated with better treatment outcomes compared with shorter durations of treatment or no treatment.81 However, neither we northe FDA78 recommend that treatment retention be a stand-alone clinical end point, as retention can be easily influenced or driven by factors external to the intervention being examined. We recommend that at least 1 outcome consider general patient functioning as assessed through pre-post changes in DSM OUD diagnostic status or symptom criteria,82,83 quality of life assessment tools, or other patient-centered outcomes that can better capture how a treatment is affecting a patient’s life beyond acute opioid exposure.79

Secondary Outcomes

Key secondary outcomes, which could be primary outcomes depending on the aims of the study, include: (1) opioid withdrawal signs and symptoms; (2) opioidcraving; (3) treatment adherence; (4) treatment satisfaction; (5) physical health (eg, comorbid diagnoses, including chronicpain); (6) mental health (eg, anxiety, depression, and other substance use); (7) cognitive and physical functioning (eg, memory, attention, sleep duration and quality, and pain severity); (8) personal and social functioning (eg, family and social relations, criminal behavior, employment, schooling, relationships, and housing and food stability); (9) health risk behavior (eg, hospitalizations, overdoses), and (10) risk of medication misuse (eg, rewarding or reinforcing effects of medication).

Risk and Adverse Events

A critical outcome in OUD trials includes opioid-related overdose or death, which is at increased risk during treatment initiation and the first several weeks after initiating abstinence or attempting opioid withdrawal.84,85 We recommend that trials, especially early treatment trials, include frequent assessment of these opioid-related adverse events, which include hospitalization, naloxone administration, and emergency department visits. Trials should also include counseling on opioid overdose risk knowledge at the onset of enrollment (eg, Brief Opioid Overdose Knowledge tutorial86 or the Overdose Education and Naloxone Distribution training) and provide naloxone.

Additional opioid-specific risks that might be monitored include infectious disease exposure and seroconversion rates (eg, HIV and hepatitis C). Emerging treatments may have unique adverse effects and events that should be monitored. For example, immunotherapeutics, such as vaccines and monoclonal antibodies specific for opioids, should be carefully evaluated for immune-related adverse effects in immunocompromised patients.42 In contrast, some sedative-hypnotic medications and cannabinoids have risks, including acute psychiatric and/or physical health consequences, misuse risk, drug-drug interactions, and diversion that should be monitored.87,88 Examples of potential risks of emerging treatments covered in the present review are included in the Table.

Challenges and Opportunities

Regulatory requirements and quality control issues, including variations in regulation at the regional and national levels in the US and other countries, can make large-scale clinical trials challenging. For example, cannabis (and other cannabinoids) and psilocybin (and other psychedelics) are all classified as schedule I drugs according to the Federal US Controlled Substances Act (ie, drugs with no currently accepted medical use and a high potential for misuse), making it more challenging and administratively burdensome to conduct clinical trials. Relatedly, both classes of drugs have a controversial history, including issues with social acceptance and legality.89 Meanwhile, state-level regulation of cannabinoids has led to variable (if any) manufacturing standards across states, resulting in intervariations and intravariations in potency and dosing across cannabinoid products. This makes it difficult to generalize research findings across some marketed consumer products.

These challenges and perspectives are slowly changing, as evidenced by the recent FDA breakthrough therapy designation for psilocybin in the treatment of depression, and 3,4-methylenedioxy-methamphetamine (MDMA) in the treatment of posttraumatic stress disorder.90 In contrast, opioid vaccines are not designated as controlled substances by the US Drug Enforcement Administration (DEA), and therefore, DEA regulations would not complicate treatment per se. However, opioid conjugate vaccines consist of multiple components, including an opioid-based small molecule hapten, which could be regulated by the DEA as either a schedule I or II drug, thereby affecting research and manufacturing.91 Manufacturing challenges related to DEA drug scheduling apply to a broad range of compounds currently in development, including synthetic cannabinoids, psychedelics, and nontraditional opioid receptor agonists and antagonists.

Another challenge is that the types of opioids being used has expanded from commercially produced opioids and heroin to also include fentanyl and/or its structural analogs, resulting in a dynamic opioid marketplace for which research may lag street-level use, type of drug, and availability. Recent data suggest increased exposure to fentanyl and its structural analogs across the US.92,93 Opioids produce diverse effects on the development and nature of opioid physical dependence and withdrawal, and fentanyl appears to be engendering a unique and particularly severe withdrawal syndrome. Establishing a treatment’s efficacy becomes especially challenging when the type of substance being targeted has such wide variability in terms of potency, route of administration, detectability, and potential for adverse outcomes.

A third challenge is that the complexity of OUD and its different stages of development are likely to have different (albeit over-lapping) underlying mechanisms that require different types of or combinations of treatments.11 For example, early sporadic use is a different stage in the life cycle from years of chronic, daily use. Furthermore, medication alone is often not a sufficient treatment for OUD, and it is important to include psychosocial and behavioral interventions and to tailor these nonpharmacological interventions to the stage of opioid use. There remain gaps in our understanding of how best to combine pharmacological and behavioral treatments.58

Despite these and other challenges, there are valuable opportunities for clinical trials with emerging treatments. Research methods are developing quickly, especially in sleep measurement, wearable devices for drug detection, remote data collection (eg, telehealth and wearable technology), and the development of genetic bio-markers for selection of phenotypes and endophenotypes that may better reflect underlying neurobiological mechanisms. The present review focused on study design considerations for clinical trials and did not discuss other relevant types of research, including pre-clinical studies, laboratory-based within-subject human studies, and observational/epidemiological studies.

Conclusions

The Box provides a summary of the key considerations and recommendations for clinical trials evaluating emerging non-MOR treatments for OUD. Promoting a unifying structure of best research practices as described in the present review will help the field build consensus as to the appropriate methodological strategies and prevent otherwise promising targets from languishing or being abandoned because of problematic study designs rather than true lack of efficacy or lack of uptake. In the context of a continually evolving and escalating opioid crisis, research must prioritize both innovation and efficiency. The field and the patients with whom we work will be best served by maintaining an open dialogue to develop a consistent methodological framework for the assessment and treatment of OUD.

Box. Key Recommendations and Considerations.

Study Objectives

  • Prospective trial registration prior to the start data collection in publicly accessible database, including primary and secondary outcomes, hypotheses, and study objectives.

  • Priority should be given to specifying the stage in OUD treatment that will be targeted with the intervention (eg, current active use of opioids, acute abstinence, nonmedically supervised withdrawal, and/or supervised withdrawal, early recovery, or long-term recovery) and determining whether the emerging treatment will be adjunctive to or independent of existing OUD treatments.

Clinical Trial Design

  • Study design will ideally be double-blind randomized clinical trial.

  • Comparators should include a placebo group (when ethically appropriate) and/or an active control comparison(s).

  • If the novel treatment is a stand-alone intervention, then comparison should include an existing, evidence-based OUD treatment (eg, methadone, buprenorphine, naltrexone, or behavioral/psychosocial support).

Sample

  • Participants should be a representative, diverse population of patients (ie, age, sex, sexual orientation, race and ethnicity, socioeconomic status, and history of substance use).

  • Exclusion criteria that are too restrictive and may negatively affect the generalizability of the study should be carefully evaluated and included on the basis of safety or another enhanced rationale considered (eg, exclusion of participants with concurrent medical, physical, or mental health issues).

Primary End Point

  • Primary outcomes should be chosen to align with the study objectives and the phase of treatment that is to be targeted (eg, symptoms of opioid withdrawal or craving will be more important to measure in early recovery rather than during long-term recovery). In addition, primary outcomes will need to be tailored to the expected treatment indication (eg, sleep measures for a sleep intervention).

  • At minimum, we recommend that primary outcomes for trials beyond phase I include opioid use behavior, treatment retention, and at least 1 outcome that addresses global functioning (eg, change in DSM criteria, quality of life).

  • A dichotomous measure to define responder (based on opioid abstinence or reduction in opioid use) should be a primary outcome, but also consider continuous measures of opioid use (ie, quantity, frequency).

  • Selection of end points should be informed by input from patients and family members to determine the most salient OUD symptoms/experiences and outcomes.

Secondary Outcomes

  • Potential secondary outcomes should include opioid withdrawal and/or craving, treatment adherence and satisfaction, physical and mental health, risk of misuse of study intervention, patient-focused outcomes, such as psychosocial functioning (including employment and legal issues), sleep, pain, and cognitive functioning, and health outcomes (eg, viral load if positive for HIV or hepatitis C virus).

Assessment of Harms

  • Adverse events, including opioid-related adverse events (eg, hospitalization, naloxone administration, visits to emergency department), and reasons for premature terminations from trial should be collected and carefully reviewed with sensitivity to relapse risk and overdose.

Abbreviation: OUD, opioid use disorder.

Funding/Support:

Financial support for this article was provided by the Analgesic, Anesthetic, and Addiction Clinical Trial Translations, Innovations, Opportunities, and Networks (ACTTION) public-private partnership with the US Food and Drug Administration (grant U01-FD005936).

Role of the Funder/Sponsor:

The Analgesic, Anesthetic, and Addiction Clinical Trial Translations, Innovations, Opportunities, and Networks (ACTTION) public-private partnership had a role in the preparation and review of the manuscript and decision to submit the manuscript for publication, and the ACTTION Executive Committee approved the manuscript.

Conflict of Interest Disclosures:

Dr Kiluk has received grants from the National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism and personal fees from the Analgesic, Anesthetic, and Addiction Clinical Trial Translations, Innovations, Opportunities, and Networks (ACTTION) during the conduct of the study as well as personal fees from CBT4CBT and Sparian Biosciences outside the submitted work. Dr Kleykamp has received personal fees from ACTTION during the conduct of the study; personal fees from American Society of Addiction Medicine, Pinney Associates, Symplr, ECRI, STATinMED, and Palladian Associates; and honoraria from Filter Magazine and Next Avenue outside the submitted work. Dr Comer has received grants from Alkermes, Lyndra Therapeutics, IntraCellular Therapies, GoMedical, BioXcel, and Janssen as well as consulting fees from Mallinckrodt, Alkermes, Clinilabs, Opiant, Nektar, and Otsuka outside the submitted work. Dr Griffiths has received grants from the Steven and Alexandra Cohen Foundation and from Tim Ferris, Matt Mullenweg, Blake Mycoskie, and Craig Nerenberg during the conduct of the study as well as personal fees from the Heffter Research Institute outside the submitted work. Dr Johnson has received grants from the National Institutes of Health, the Heffter Research Institute, and Mydecine Innovations Group; personal fees from AJNA Labs, AWAKN Life Sciences, Beckley Psychedelic, Entheogen Biomedical, Mind Medicine, Field Trip Psychedelics, Silo Pharma, and Otsuka Pharmaceutical Development and Commercialization; and nonfinancial support from Mydecine Innovations Group outside the submitted work. Dr Kampman has received grants from US World Meds and Alkermes outside the submitted work. Dr Pravetoni has received grants from the National Institutes of Health during the conduct of the study and has a patent pending for disclosing technology related to vaccines and monoclonal antibodies and other treatment strategies for substance use disorders and overdose. Dr Vandrey has received personal fees from Canopy Health Innovations, MyMD Pharmaceuticals, Mira1a Pharmaceuticals, WebMD, Radicle Science, and Syqe Medical during the conduct of the study. Dr Bergeria has received grants from the National Institute on Drug Abuse and ACTTION during the conduct of the study as well as grants from Canopy Growth Corporation outside the submitted work. Dr Bogenschutz has received research funding from the National Institute on Alcohol Abuse and Alcoholism, National Institute on Drug Abuse, Heffter Research Institute, Mind Medicine, Tilray Canada, Multidisciplinary Association for Psychedelic Studies (MAPS), B.More, Turnbull Family Foundation, Fournier Family Foundation, Dr Bronner’s Family Foundation, and Riverstyx Foundation and from Rodrigo Nino, a philanthropic donor, as well as personal fees from the Heffter Research Institute, Ajna Labs, Beckley Psytech, Journey Colab, and Bright Minds Biosciences outside the submitted work. Dr Brown has received grants from the National Institutes of Health, Health Services and Resources Administration, Substance Abuse and Mental Health Services Administration, Usona Institute, Multi-Disciplinary Association for Psychedelic Studies, Etheridge Foundation, and Heffter Research Institute outside the submitted work. Dr Dunn has received grants from the National Institutes of Health as well as personal fees from Mind Medicine, Canopy Corporation, and Canopy-Beckley outside the submitted work. Dr Dworkin has received grants from US Food and Drug Administration and National Institutes of Health during the conduct of the study; personal fees from Abide, Acadia, Adynxx, Analgesic Solutions, Aptinyx, Aquinox, Asahi Kasei, Astellas, Beckley, Biogen, Biohaven, Biosplice, Boston Scientific, Braeburn, Cardialen, Celgene, Centrexion, Chiesi, Chromocell, Clexio, Collegium, Concert, Confo, Decibel, Editas, Eli Lilly, Endo, Eupraxia, Exicure, Glenmark, Gloriana, Grace, Hope, Lotus, Mainstay, Merck, Mind Medicine, Neumentum, Neurana, NeuroBo, Novaremed, Novartis, Oxford Cannabinoid Technologies, OliPass, Pfizer, Q-State, Reckitt Benckiser, Regenacy, Sangamo, Sanifit, Scilex, Semnur, SIMR Biotech, Sinfonia, SK Biopharmaceuticals, Sollis, SPRIM, Teva, Theranexus, Toray, Vertex, Vizuri, and WCG and holds equity in Ethismos, Regenacy, and Mind Medicine outside the submitted work. Dr Finan has received personal fees from Ninnion Therapeutics outside the submitted work. Dr Hendricks has received grants from the Heffter Research Institute and National Institutes of Health as well as personal fees from Bright Minds Biosciences, Eleusis Health Solutions, Reset Pharmaceuticals, and Silo Pharma outside the submitted work. Dr Kosten has received grants from the US Department of Defense, National Institutes of Health, Toomim Family Foundation, and John O’Quinn Foundation as well as personal fees from Alkermes, Amygdala Neurosciences, Aptinyx, Arbor Pharma, Astellas, BioXcel, Boehringer Ingelheim, Braeburn, Data Analytics, Diamond, Embera, Epiodyne, Indivior, Intra-Cellular Therapies, Mallinckrodt, Medscape, Nektar, Nirsum, Novartis, Opiant, Otsuka, Pfizer, Phenex, Promentis, Relmada, Sage, United Neuroscience, US World Meds, and Zynerba. Dr Levin has received grants from US World Meds Medication, Substance Abuse and Mental Health Services Administration, and National Institute on Drug Abuse; personal fees from Major League Baseball; and nonfinancial support from Indivior outside the submitted work; and served on the scientific advisory boards of Alkermes, Indivior, Novartis, Teva, and US World Meds. Dr Raison has received personal fees from Usona Institute, Novartis, Alfasigma, Emory Healthcare, and Otsuka during the conduct of the study. Dr Rasmussen was Director of the Division of Therapeutics and Medical Consequences of the National Institute on Drug Abuse from 2018 to 2021. Dr Turk has received personal fees from GlaxoSmithKline/Novartis and nonfinancial support from Pfizer outside the submitted work and is Editor-in-Chief of the Clinical Journal of Pain and Associate Director of ACTTION. Dr Weiss has received personal fees from Alkermes, Cerevel Therapeutics, Takeda Pharmaceuticals, and Astellas Pharma outside the submitted work. Dr Strain has received personal fees from Caron, Cerevel, Elsevier, Fast Track Drugs and Biologics, Otsuka, Pear Therapeutics, and UpToDate as well as nonfinancial support from Masimo/Innovative Health Solutions outside the submitted work. No other disclosures were reported.

Footnotes

Disclaimer: The views expressed in this article are those of the authors and no official endorsement by the US Food and Drug Administration or the pharmaceutical and device companies that provided unrestricted grants to support the activities of the Analgesic, Anesthetic, and Addiction Clinical Trial Translations, Innovations, Opportunities, and Networks (ACTTION) public-private partnership should be inferred.

REFERENCES

  • 1.Volkow ND, Woodcock J, Compton WM, et al. Medication development in opioid addiction: meaningful clinical end points. Sci Transl Med. 2018; 10(434):eaan2595. doi: 10.1126/scitranslmed.aan2595 [DOI] [PubMed] [Google Scholar]
  • 2.Degenhardt L, Charlson F, Mathers B, et al. The global epidemiology and burden of opioid dependence: results from the Global Burden of Disease 2010 study. Addiction. 2014;109(8):1320–1333. doi: 10.1111/add.12551 [DOI] [PubMed] [Google Scholar]
  • 3.Hser Y-I, Mooney LJ, Saxon AJ, et al. High mortality among patients with opioid use disorder in a large healthcare system. J Addict Med. 2017;11 (4):315–319. doi: 10.1097/ADM.0000000000000312 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Volkow ND, Blanco C. Medications for opioid use disorders: clinical and pharmacological considerations. J Clin Invest. 2020;130(1):10–13. doi: 10.1172/JCI134708 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.National Academies of Sciences, Engineering, and Medicine; Health and Medicine Division; Board on Health Sciences Policy; Committee on Medication-Assisted Treatment for Opioid Use Disorder; Mancher M, Leshner AI, eds. Medications for Opioid Use Disorder Save Lives. National Academies Press; 2019. [PubMed] [Google Scholar]
  • 6.Morgan JR, Schackman BR, Leff JA, Linas BP, Walley AY. Injectable naltrexone, oral naltrexone, and buprenorphine utilization and discontinuation among individuals treated for opioid use disorder in a United States commercially insured population. J Subst Abuse Treat. 2018;85:90–96. doi: 10.1016/j.jsat.2017.07.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Lee JD, Nunes EV Jr, Novo P, et al. Comparative effectiveness of extended-release naltrexone versus buprenorphine-naloxone for opioid relapse prevention (X:BOT): a multicentre, open-label, randomised controlled trial. Lancet. 2018;391 (10118):309–318. doi: 10.1016/S0140-6736(17)32812-X [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.O’Connor AM, Cousins G, Durand L, Barry J, Boland F. Retention of patients in opioid substitution treatment: a systematic review. PLoS One. 2020;15(5):e0232086. doi: 10.1371/journal.pone.0232086 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Volkow ND, Frieden TR, Hyde PS, Cha SS. Medication-assisted therapies–tackling the opioid-overdose epidemic. N Engl J Med. 2014;370 (22):2063–2066. doi: 10.1056/NEJMp1402780 [DOI] [PubMed] [Google Scholar]
  • 10.Fuehrlein BS, Ross DA. Opioid use disorder: a desperate need for novel treatments. Biol Psychiatry. 2017;81(7):e43–e45. doi: 10.1016/j.biopsych.2017.01.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Rasmussen K, White DA, Acri JB. NIDA’s medication development priorities in response to the opioid crisis: ten most wanted. Neuropsychopharmacology. 2019;44(4):657–659. doi: 10.1038/s41386-018-0292-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Blanco C, Volkow ND. Management of opioid use disorder in the USA: present status and future directions. Lancet. 2019;393(10182):1760–1772. doi: 10.1016/S0140-6736(18)33078-2 [DOI] [PubMed] [Google Scholar]
  • 13.Strain EC, Kampman KM, Weiss RD. Moving beyond medications that act at the μ receptor in the treatment of opioid use disorder. JAMA Psychiatry. 2021;78(7):701–702. doi: 10.1001/jamapsychiatry.2021.0259 [DOI] [PubMed] [Google Scholar]
  • 14.Analgesic, Anesthetic, and Addiction Clinical Trial Translations, Innovations, Opportunities, and Networks. Consortium for Addiction Research on Efficacy and Safety (CARES). Accessed June 10, 2022. https://www.acttion.org/cares [Google Scholar]
  • 15.Food US and Administration Drug. Patient-focused drug development: collecting comprehensive and representative input. Accessed July 27, 2021. https://www.fda.gov/regulatory-information/search-fda-guidance-documents/patient-focused-drug-development-collecting-comprehensive-and-representative-input
  • 16.Dennis BB, Sanger N, Bawor M, et al. A call for consensus in defining efficacy in clinical trials for opioid addiction: combined results from a systematic review and qualitative study in patients receiving pharmacological assisted therapy for opioid use disorder. Trials. 2020;21(1):30. doi: 10.1186/s13063-019-3995-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Cook NS, Cave J, Holtorf A-P. Patient preference studies during early drug development: aligning stakeholders to ensure development plans meet patient needs. Front Med (Lausanne). 2019;6:82. doi: 10.3389/fmed.2019.00082 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Wallerstein N, Duran B. Community-based participatory research contributions to intervention research: the intersection of science and practice to improve health equity. Am J Public Health. 2010; 100(suppl 1):S40–S46. doi: 10.2105/AJPH.2009.184036 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Sheridan S, Schrandt S, Forsythe L, Hilliard TS, Paez KA; Advisory Panel on Patient Engagement (2013 Inaugural Panel). The PCORI Engagement Rubric: promising practices for partnering in research. Ann Fam Med. 2017;15(2):165–170. doi: 10.1370/afm.2042 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Humphreys K, Saitz R. Should physicians recommend replacing opioids with cannabis? JAMA. 2019;321(7):639–640. doi: 10.1001/jama.2019.0077 [DOI] [PubMed] [Google Scholar]
  • 21.Hurd YL. Cannabidiol: swinging the marijuana pendulum from ‘weed’ to medication to treat the opioid epidemic. Trends Neurosci. 2017;40(3):124–127. doi: 10.1016/j.tins.2016.12.006 [DOI] [PubMed] [Google Scholar]
  • 22.Hurd YL, Yoon M, Manini AF, et al. Early phase in the development of cannabidiol as a treatment for addiction: opioid relapse takes initial center stage. Neurotherapeutics. 2015;12(4):807–815. doi: 10.1007/s13311-015-0373-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Lucas P Rationale for cannabis-based interventions in the opioid overdose crisis. Harm Reduct J. 2017;14(1):58. doi: 10.1186/s12954-017-0183-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Bisaga A, Sullivan MA, Glass A, et al. The effects of dronabinol during detoxification and the initiation of treatment with extended release naltrexone. Drug Alcohol Depend. 2015;154:38–45. doi: 10.1016/j.drugalcdep.2015.05.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Jicha CJ, Lofwall MR, Nuzzo PA, Babalonis S, Elayi SC, Walsh SL. Safety of oral dronabinol during opioid withdrawal in humans. Drug Alcohol Depend. 2015;157:179–183. doi: 10.1016/j.drugalcdep.2015.09.031 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Lofwall MR, Babalonis S, Nuzzo PA, Elayi SC, Walsh SL. Opioid withdrawal suppression efficacy of oral dronabinol in opioid dependent humans. Drug Alcohol Depend. 2016;164:143–150. doi: 10.1016/j.drugalcdep.2016.05.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Wiese B, Wilson-Poe AR. Emerging evidence for cannabis’ role in opioid use disorder. Cannabis Cannabinoid Res. 2018;3(1):179–189. doi: 10.1089/can.2018.0022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Nichols DE, Johnson MW, Nichols CD. Psychedelics as medicines: an emerging new paradigm. Clin Pharmacol Ther. 2017;101(2):209–219. doi: 10.1002/cpt.557 [DOI] [PubMed] [Google Scholar]
  • 29.Garcia-Romeu A, Kersgaard B, Addy PH. Clinical applications of hallucinogens: a review. Exp Clin Psychopharmacol. 2016;24(4):229–268. doi: 10.1037/pha0000084 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Krebs TS, Johansen PØ. Lysergic acid diethylamide (LSD) for alcoholism: meta-analysis of randomized controlled trials. J Psychopharmacol. 2012;26(7):994–1002. doi: 10.1177/0269881112439253 [DOI] [PubMed] [Google Scholar]
  • 31.Garcia-Romeu A, Davis AK, Erowid E, Erowid F, Griffiths RR, Johnson MW. Persisting reductions in cannabis, opioid, and stimulant misuse after naturalistic psychedelic use: an online survey. Front Psychiatry. 2020;10:955–955. doi: 10.3389/fpsyt.2019.00955 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Pisano VD, Putnam NP, Kramer HM, Franciotti KJ, Halpern JH, Holden SC. The association of psychedelic use and opioid use disorders among illicit users in the United States. J Psychopharmacol. 2017;31(5):606–613. doi: 10.1177/0269881117691453 [DOI] [PubMed] [Google Scholar]
  • 33.Belouin SJ, Henningfield JE. Psychedelics: where we are now, why we got here, what we must do. Neuropharmacology. 2018;142:7–19. doi: 10.1016/j.neuropharm.2018.02.018 [DOI] [PubMed] [Google Scholar]
  • 34.Argento E, Tupper KW, Socias ME. The tripping point: the potential role of psychedelic-assisted therapy in the response to the opioid crisis. Int J Drug Policy. 2019;66:80–81. doi: 10.1016/j.drugpo.2018.11.006 [DOI] [PubMed] [Google Scholar]
  • 35.Cheatle MD, Webster LR. Opioid therapy and sleep disorders: risks and mitigation strategies. Pain Med. 2015;16(suppl 1):S22–S26. doi: 10.1111/pme.12910 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Huhn AS, Harris J, Cleveland HH, et al. Ecological momentary assessment of affect and craving in patients in treatment for prescription opioid dependence. Brain Res Bull. 2016;123:94–101. doi: 10.1016/j.brainresbull.2016.01.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Lydon-Staley DM, Cleveland HH, Huhn AS, et al. Daily sleep quality affects drug craving, partially through indirect associations with positive affect, in patients in treatment for nonmedical use of prescription drugs. Addict Behav. 2017;65:275–282. doi: 10.1016/j.addbeh.2016.08.026 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Fathi HR, Yoonessi A, Khatibi A, Rezaeitalab F, Rezaei-Ardani A. Crosstalk between sleep disturbance and opioid use disorder: a narrative review. Addict Health. 2020;12(2):140–158. doi: 10.22122/ahj.v12i2.249 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Freeman LK, Gottfredson NC. Using ecological momentary assessment to assess the temporal relationship between sleep quality and cravings in individuals recovering from substance use disorders. Addict Behav. 2018;83:95–101. doi: 10.1016/j.addbeh.2017.11.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Dolsen MR, Harvey AG. Life-time history of insomnia and hypersomnia symptoms as correlates of alcohol, cocaine and heroin use and relapse among adults seeking substance use treatment in the United States from 1991 to 1994. Addiction. 2017;112(6):1104–1111. doi: 10.1111/add.13772 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.James MH, Fragale JE, Aurora RN, Cooperman NA, Langleben DD, Aston-Jones G. Repurposing the dual orexin receptor antagonist suvorexant for the treatment of opioid use disorder: why sleep on this any longer? Neuropsychopharmacology. 2020;45 (5):717–719. doi: 10.1038/s41386-020-0619-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Pravetoni M, Comer SD. Development of vaccines to treat opioid use disorders and reduce incidence of overdose. Neuropharmacology. 2019; 158:107662. doi: 10.1016/j.neuropharm.2019.06.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Baehr C, Pravetoni M. Vaccines to treat opioid use disorders and to reduce opioid overdoses. Neuropsychopharmacology. 2019;44(1):217–218. doi: 10.1038/s41386-018-0197-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Bremer PT, Kimishima A, Schlosburg JE, Zhou B, Collins KC, Janda KD. Combatting synthetic designer opioids: a conjugate vaccine ablates lethal doses of fentanyl class drugs. Angew Chem Int Ed Engl. 2016;55(11):3772–3775. doi: 10.1002/anie.201511654 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Ohia-Nwoko O, Kosten TA, Haile CN. Animal models and the development of vaccines to treat substance use disorders. Int Rev Neurobiol. 2016; 126:263–291. doi: 10.1016/bs.irn.2016.02.009 [DOI] [PubMed] [Google Scholar]
  • 46.Tenney RD, Blake S, Bremer PT, et al. Vaccine blunts fentanyl potency in male rhesus monkeys. Neuropharmacology. 2019;158:107730. doi: 10.1016/j.neuropharm.2019.107730 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Townsend EA, Blake S, Faunce KE, et al. Conjugate vaccine produces long-lasting attenuation of fentanyl vs. food choice and blocks expression of opioid withdrawal-induced increases in fentanyl choice in rats. Neuropsychopharmacology. 2019;44(10):1681–1689. doi: 10.1038/s41386-019-0385-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Banks ML, Olson ME, Janda KD. Immunopharmacotherapies for treating opioid use disorder. Trends Pharmacol Sci. 2018;39(11):908–911. doi: 10.1016/j.tips.2018.08.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Raleigh MD, King SJ, Baruffaldi F, et al. Pharmacological mechanisms underlying the efficacy of antibodies generated by a vaccine to treat oxycodone use disorder. Neuropharmacology. 2021;195:108653. doi: 10.1016/j.neuropharm.2021.108653 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Baehr C, Kelcher AH, Khaimraj A, et al. Monoclonal antibodies counteract opioid-induced behavioral and toxic effects in mice and rats. J Pharmacol Exp Ther. 2020;375(3):469–477. doi: 10.1124/jpet.120.000124 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Savage C, McCabe OL. Residential psychedelic (LSD) therapy for the narcotic addict. a controlled study. Arch Gen Psychiatry. 1973;28(6):808–814. doi: 10.1001/archpsyc.1973.01750360040005 [DOI] [PubMed] [Google Scholar]
  • 52.Stein MD, Kurth ME, Sharkey KM, Anderson BJ, Corso RP, Millman RP. Trazodone for sleep disturbance during methadone maintenance: a double-blind, placebo-controlled trial. Drug Alcohol Depend. 2012;120(1–3):65–73. doi: 10.1016/j.drugalcdep.2011.06.026 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Akbarzadeh A, Norouzian D, Farhangi A, et al. Immunotherapy of 347 volunteer outpatient morphine addicts by human therapeutic morphine vaccine in Kermanshah Province of Iran. J Pharmacol Toxicol. 2009;4:30–35. doi: 10.3923/jpt.2009.30.35 [DOI] [Google Scholar]
  • 54.Clinical Trials of Multivalent Opioid Vaccine Components. ClinicalTrials.gov identifier: NCT04458545. Updated July 5, 2022. Accessed August 20, 2022. https://clinicaltrials.gov/ct2/show/NCT04458545
  • 55.Ekhtiari H, Tavakoli H, Addolorato G, et al. Transcranial electrical and magnetic stimulation (tES and TMS) for addiction medicine: a consensus paper on the present state of the science and the road ahead. Neurosci Biobehav Rev. 2019;104:118–140. doi: 10.1016/j.neubiorev.2019.06.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Ford I, Norrie J. Pragmatic trials. N Engl J Med. 2016;375(5):454–463. doi: 10.1056/NEJMra1510059 [DOI] [PubMed] [Google Scholar]
  • 57.Pallmann P, Bedding AW, Choodari-Oskooei B, et al. Adaptive designs in clinical trials: why use them, and how to run and report them. BMC Med. 2018;16(1):29. doi: 10.1186/s12916-018-1017-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Dugosh K, Abraham A, Seymour B, McLoyd K, Chalk M, Festinger D. A systematic review on the use of psychosocial interventions in conjunction with medications for the treatment of opioid addiction. J Addict Med. 2016;10(2):93–103. doi: 10.1097/ADM.0000000000000193 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Mauri L, D’Agostino RB Sr. Challenges in the design and interpretation of noninferiority trials. N Engl J Med. 2017;377(14):1357–1367. doi: 10.1056/NEJMra1510063 [DOI] [PubMed] [Google Scholar]
  • 60.Uscher-Pines L, Huskamp HA, Mehrotra A. Treating patients with opioid use disorder in their homes: an emerging treatment model. JAMA. 2020;324(1):39–40. doi: 10.1001/jama.2020.3940 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Ho C, Argáez C. Telehealth-Delivered Opioid Agonist Therapy for the Treatment of Adults With Opioid Use Disorder: Review of Clinical Effectiveness, Cost-effectiveness, and Guidelines. Canadian Agency for Drugs and Technologies in Health; 2018. [PubMed] [Google Scholar]
  • 62.Dahne J, Tomko RL, McClure EA, Obeid JS, Carpenter MJ. Remote methods for conducting tobacco-focused clinical trials. Nicotine Tob Res. 2020;22(12):2134–2140. doi: 10.1093/ntr/ntaa105 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Crowley D, Delargy I. A national model of remote care for assessing and providing opioid agonist treatment during the COVID-19 pandemic: a report. Harm Reduct J. 2020;17(1):49. doi: 10.1186/s12954-020-00394-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Mitchell MM, Mendelson J, Gryczynski J, Carswell SB, Schwartz RP. A novel telehealth platform for alcohol use disorder treatment: preliminary evidence of reductions in drinking. Am J Drug Alcohol Abuse. 2020;46(3):297–303. doi: 10.1080/00952990.2019.1658197 [DOI] [PubMed] [Google Scholar]
  • 65.Korthuis PT, McCarty D, Weimer M, et al. Primary care–based models for the treatment of opioid use disorder: a scoping review. Ann Intern Med. 2017;166(4):268–278. doi: 10.7326/M16-2149 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Donroe JH, Bhatraju EP, Tsui JI, Edelman EJ. Identification and management of opioid use disorder in primary care: an update. Curr Psychiatry Rep. 2020;22(5):23. doi: 10.1007/s11920-020-01149-0 [DOI] [PubMed] [Google Scholar]
  • 67.Hendricks PS, Copes H, Family N, Williams LT, Luke D, Raz S. Perceptions of safety, subjective effects, and beliefs about the clinical utility of lysergic acid diethylamide in healthy participants within a novel intervention paradigm: qualitative results from a proof-of-concept study. J Psychopharmacol. 2022;36(3):337–347. doi: 10.1177/02698811211055855 [DOI] [PubMed] [Google Scholar]
  • 68.Family N, Hendricks PS, Williams LT, et al. Safety, tolerability, pharmacokinetics, and subjective effects of 50, 75, and 100 μg LSD in healthy participants within a novel intervention paradigm: a proof-of-concept study. J Psychopharmacol. 2022;36(3):321–336. doi: 10.1177/02698811211069103 [DOI] [PubMed] [Google Scholar]
  • 69.Priester MA, Browne T, Iachini A, Clone S, DeHart D, Seay KD. Treatment access barriers and disparities among individuals with co-occurring mental health and substance use disorders: an integrative literature review. J Subst Abuse Treat. 2016;61:47–59. doi: 10.1016/j.jsat.2015.09.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Olfson M, Zhang VS, Schoenbaum M, King M. Trends in buprenorphine treatment in the United States, 2009–2018. JAMA. 2020;323(3):276–277. doi: 10.1001/jama.2019.18913 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Gordon AJ, Drexler K, Hawkins EJ, et al. Stepped Care for Opioid Use Disorder Train the Trainer (SCOUTT) initiative: expanding access to medication treatment for opioid use disorder within Veterans Health Administration facilities. Subst Abus. 2020;41(3):275–282. doi: 10.1080/08897077.2020.1787299 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Short VL, Hand DJ, MacAfee L, Abatemarco DJ, Terplan M. Trends and disparities in receipt of pharmacotherapy among pregnant women in publically funded treatment programs for opioid use disorder in the United States. J Subst Abuse Treat. 2018;89:67–74. doi: 10.1016/j.jsat.2018.04.003 [DOI] [PubMed] [Google Scholar]
  • 73.Eghaneyan BH, Sanchez K, Haeny AM, et al. Hispanic participants in the National Institute on Drug Abuse’s Clinical Trials Network: a scoping review of two decades of research. Addict Behav Rep. 2020;12:100287. doi: 10.1016/j.abrep.2020.100287 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Pinedo M A current re-examination of racial/ethnic disparities in the use of substance abuse treatment: do disparities persist? Drug Alcohol Depend. 2019;202:162–167. doi: 10.1016/j.drugalcdep.2019.05.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Abraham AJ, Andrews CM, Yingling ME, Shannon J. Geographic disparities in availability of opioid use disorder treatment for Medicaid enrollees. Health Serv Res. 2018;53(1):389–404. doi: 10.1111/1475-6773.12686 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Stahler GJ, Mennis J. Treatment outcome disparities for opioid users: are there racial and ethnic differences in treatment completion across large US metropolitan areas? Drug Alcohol Depend. 2018;190:170–178. doi: 10.1016/j.drugalcdep.2018.06.006 [DOI] [PubMed] [Google Scholar]
  • 77.Biondi BE, Zheng X, Frank CA, Petrakis I, Springer SA. A literature review examining primary outcomes of medication treatment studies for opioid use disorder: what outcome should be used to measure opioid treatment success? Am J Addict. 2020;29(4):249–267. doi: 10.1111/ajad.13051 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.US Food and Drug Administration. Opioid use disorder: endpoints for demonstrating effectiveness of drugs for treatment guidance for industry. Accessed July 27, 2021. https://www.fda.gov/regulatory-information/search-fda-guidance-documents/opioid-use-disorder-endpointsdemonstrating-effectiveness-drugs-treatment-guidance-industry
  • 79.Kiluk BD, Fitzmaurice GM, Strain EC, Weiss RD. What defines a clinically meaningful outcome in the treatment of substance use disorders: reductions in direct consequences of drug use or improvement in overall functioning? Addiction. 2019;114(1):9–15. doi: 10.1111/add.14289 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Fitzmaurice GM, Lipsitz SR, Weiss RD. Within-treatment frequency of use versus abstinence as a predictor of longitudinal post-treatment follow-up assessments of drug use. Drug Alcohol Depend. 2020;208:107857. doi: 10.1016/j.drugalcdep.2020.107857 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Timko C, Schultz NR, Cucciare MA, Vittorio L, Garrison-Diehn C. Retention in medication-assisted treatment for opiate dependence: a systematic review. J Addict Dis. 2016;35(1):22–35. doi: 10.1080/10550887.2016.1100960 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Marsden J, Tai B, Ali R, Hu L, Rush AJ, Volkow N. Measurement-based care using DSM-5 for opioid use disorder: can we make opioid medication treatment more effective? Addiction. 2019;114(8):1346–1353. doi: 10.1111/add.14546 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Kiluk BD, Frankforter TL, Cusumano M, Nich C, Carroll KM. Change in DSM-5 alcohol use disorder criteria count and severity level as a treatment outcome indicator: results from a randomized trial. Alcohol Clin Exp Res. 2018. doi: 10.1111/acer.13807 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Cousins G, Teljeur C, Motterlini N, McCowan C, Dimitrov BD, Fahey T. Risk of drug-related mortality during periods of transition in methadone maintenance treatment: a cohort study. J Subst Abuse Treat. 2011;41(3):252–260. doi: 10.1016/j.jsat.2011.05.001 [DOI] [PubMed] [Google Scholar]
  • 85.Sordo L, Barrio G, Bravo MJ, et al. Mortality risk during and after opioid substitution treatment: systematic review and meta-analysis of cohort studies. BMJ. 2017;357:j1550. doi: 10.1136/bmj.j1550 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Dunn KE, Barrett FS, Yepez-Laubach C, et al. Brief Opioid Overdose Knowledge (BOOK): a questionnaire to assess overdose knowledge in individuals who use illicit or prescribed opioids. J Addict Med. 2016;10(5):314–323. doi: 10.1097/ADM.0000000000000235 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.McLarnon ME, Monaghan TL, Stewart SH, Barrett SP. Drug misuse and diversion in adults prescribed anxiolytics and sedatives. Pharmacotherapy. 2011;31(3):262–272. doi: 10.1592/phco.31.3.262 [DOI] [PubMed] [Google Scholar]
  • 88.Hasin DS, Sarvet AL, Cerdá M, et al. US adult illicit cannabis use, cannabis use disorder, and medical marijuana laws: 1991–1992 to 2012–2013. JAMA Psychiatry. 2017;74(6):579–588. doi: 10.1001/jamapsychiatry.2017.0724 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Sellers EM, Leiderman DB. Psychedelic drugs as therapeutics: no illusions about the challenges. Clin Pharmacol Ther. 2018;103(4):561–564. doi: 10.1002/cpt.776 [DOI] [PubMed] [Google Scholar]
  • 90.Mitchell JM, Bogenschutz M, Lilienstein A, et al. MDMA-assisted therapy for severe PTSD: a randomized, double-blind, placebo-controlled phase 3 study. Nat Med. 2021;27(6):1025–1033. doi: 10.1038/s41591-021-01336-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Comer SD, Pravetoni M, Coop A, Baumann MH, Cunningham CW. Potential unintended consequences of class-wide drug scheduling based on chemical structure: a cautionary tale for fentanyl-related compounds. Drug Alcohol Depend. 2021;221:108530. doi: 10.1016/j.drugalcdep.2021.108530 [DOI] [Google Scholar]
  • 92.Palamar JJ, Ciccarone D, Rutherford C, Keyes KM, Carr TH, Cottler LB. Trends in seizures of powders and pills containing illicit fentanyl in the United States, 2018 through 2021. Drug Alcohol Depend. 2022;234:109398. doi: 10.1016/j.drugalcdep.2022.109398 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Brunetti P, Pirani F, Carlier J, Giorgetti R, Busardò FP, Lo Faro AFA. A 2017–2019 update on acute intoxications and fatalities from illicit fentanyl and analogs. J Anal Toxicol. 2021;45(6):537–554. doi: 10.1093/jat/bkaa115 [DOI] [PubMed] [Google Scholar]

RESOURCES