The ASAM National Practice Guideline for the Use of Medications in the Treatment of Addiction Involving Opioid Use ∗












From Kampman K, Jarvis M. American Society of Addiction (ASAM) national practice guidelines for the use of medications in the treatment of addiction involving opioid use. J Addict Med. 2015;9(5):358–367.





Guideline Committee Members (alpha order)





  • Sandra Comer, PhD


    Disclosure information for Guideline Committee Members, the ASAM Quality Improvement Council, and External Reviewers is available respectively in Appendices III, IV, and V.




  • Chinazo Cunningham, MD, MS



  • Marc J. Fishman, MD, FASAM



  • Adam Gordon, MD, MPH, FASAM



  • Kyle Kampman, MD, Chair



  • Daniel Langleben, MD



  • Ben Nordstrom, MD, PhD



  • David Oslin, MD



  • George Woody, MD



  • Tricia Wright, MD, MS



  • Stephen Wyatt, DO





ASAM Quality Improvement Council (alpha order)





  • John Femino, MD, FASAM



  • Margaret Jarvis, MD, FASAM, Chair



  • Margaret Kotz, DO, FASAM



  • Sandrine Pirard, MD, MPH, PhD



  • Robert J. Roose, MD, MPH



  • Alexis Geier-Horan, ASAM Staff



  • Beth Haynes, ASAM Staff



  • Penny S. Mills, MBA, ASAM, Executive Vice President





Special External Reviewer





  • Michael M. Miller, MD, FASAM, FAPA





Treatment Research Institute Technical Team Members (alpha order)





  • Amanda Abraham, PhD



  • Karen Dugosh, PhD



  • David Festinger, PhD



  • Kyle Kampman, MD, Principal Investigator



  • Keli McLoyd, JD



  • Brittany Seymour, BA



  • Abigail Woodworth, MS





Executive Summary


Purpose


The American Society of Addiction Medicine (ASAM) developed this National Practice Guideline for the Use of Medications in the Treatment of Addiction Involving Opioid Use to provide information on evidence-based treatment of opioid use disorder. (Hereafter, in this document, this National Practice Guideline will be referred to as “Practice Guideline.”)


Background


Opioid use disorder is a chronic, relapsing disease, which has significant economic, personal, and public health consequences. Many readers of this Practice Guideline may recognize the term “opioid use disorder” as it is used in the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition (DSM-5), developed by the American Psychiatric Association; others may be more familiar with the term “opioid dependence,” as used in the previous edition of the DSM. 7


The ASAM defines addiction as “a primary, chronic disease of brain reward, motivation, memory, and related circuitry,” with a “dysfunction in these circuits” being reflected in “an individual pathologically pursuing reward and/or relief by substance use and other behaviors.” In this context, the preferred term by ASAM for this serious bio-psycho-social-spiritual illness would be “addiction involving opioid use.” ASAM views addiction as a fundamental neurological disorder of “brain reward, motivation, memory, and related circuitry,” and recognizes that there are unifying features in all cases of addiction, including substance-related addiction and nonsubstance-related addiction. It is clear that a variety of substances commonly associated with addiction work on specific receptors in the nervous system and on specific neurotransmitter systems. Specific pharmacological agents used in the treatment of addiction exert their effects via their actions on specific receptors. Hence, the medications used in the treatment of addiction have specific efficacy based on their own molecular structure and the particular neurotransmitters affected by that medication. Medications developed for the treatment of addiction involving opioid use may have benefits in the treatment of addiction involving an individual’s use of other substances. For instance, naltrexone, which is approved by the US Food and Drug Administration (FDA) for the treatment of opioid dependence using DSM, Fourth Edition (DSM-IV) terminology, is also FDA-approved for the treatment of alcohol dependence as per the DSM-IV guidelines. 8


The ASAM recognizes that research is yet to be done to confirm the specificity of its conceptualization of addiction as a medical and a psychiatric illness. Both the American Medical Association (AMA), as noted in various policy and position statements, and the International Classification of Diseases (ICD), recognize addiction as both a medical and a psychiatric disorder. 111,171 ASAM encourages clinicians, researchers, educators, and policy makers to use the term “addiction” regardless of whether the patient’s condition at a given point in its natural history seems to more prominently involve opioid use, alcohol use, nicotine use, or engagement in addictive behaviors such as gambling. Given the widespread North American application of the DSM’s categorization of disorders, this Practice Guideline will, for the sake of brevity and convention, use the term “opioid use disorder.”


According to the 2013 National Survey on Drug Use and Health (NSDUH), 4.5 million individuals in the United States were current (past month), nonmedical users of prescription opioids. Nonmedical use of opioids and other prescription drugs constitute hazardous and risky behavior that should be discouraged given the potential that unauthorized use of such substances has for harm (to the user). Medication therapy related to opioids focuses not only on nonmedical use but also on an attempt to treat the medical illness, addiction. The 2013 NSDUH further found that 1.9 million persons in America met DSM-IV criteria for opioid use disorder associated with their use of prescription opioids, and that more than 0.5 million additional individuals have met DSM-IV criteria for opioid use disorder associated with their use of heroin. 146


Opioid use is associated with increased mortality. The leading causes of death in people using opioids for nonmedical purposes are overdose and trauma. 50 The injection route use (intravenous or even intramuscular [IM]) of opioids or other drugs increases the risk of being exposed to HIV, viral hepatitis, and other infectious agents.


Scope of Guideline


This Practice Guideline was developed for the evaluation and treatment of opioid use disorder and for the management of opioid overdose. The medications covered in this guideline are mainly, but not exclusively, those that have been FDA-approved for the treatment of opioid dependence, as defined in prior versions of the DSM, and not necessarily the most recent version of the manual, the DSM-5. 151 DSM-5 combined the criteria for opioid abuse and opioid dependence from prior versions of the DSM in its new diagnosis of opioid use disorder: therefore, pharmacological treatment may not be appropriate for all patients along the entire opioid use disorder continuum. In a study comparing opioid dependence from DSM-IV and opioid use disorder from DSM-5, optimal concordance occurred when four or more DSM-5 criteria were endorsed (i.e., the DSM-5 threshold for moderate opioid use disorder). 42 Other medications have been used off-label to treat opioid use disorder (clearly noted in the text); however, the Guideline Committee has not issued recommendations on the use of those medications. As a final note related to references to medications, whether FDA-approved or off-label, cost and/or cost effectiveness were not considerations in the development of this Practice Guideline.


Intended Audience


This Practice Guideline is primarily intended for clinicians involved in evaluating patients and providing authorization for pharmacological treatments at any level. The intended audience falls into the broad groups of physicians; other healthcare providers (especially those with prescribing authority); medical educators and faculty for other healthcare professionals in training; and clinical care managers, including those offering utilization management services.


Qualifying Statement


This ASAM Practice Guideline is intended to aid clinicians in their clinical decision-making and patient management. The Practice Guideline strives to identify and define clinical decision-making junctures that meet the needs of most patients in most circumstances. Clinical decision-making should involve consideration of the quality and availability of expertise and services in the community wherein care is provided. In circumstances in which the Practice Guideline is being used as the basis for regulatory or payer decisions, improvement in quality of care should be the goal. Finally, prescribed courses of treatment contained in recommendations in this Practice Guideline are effective only if the recommendations, as outlined, are followed. Because lack of patient understanding and adherence may adversely affect outcomes, clinicians should make every effort to promote the patient’s understanding of, and adherence to, prescribed and recommended pharmacological and psychosocial treatments. Patients should be informed of the risks, benefits, and alternatives to a particular treatment, and should be an active party to shared decision-making whenever feasible. Recommendations in this Practice Guideline do not supersede any federal or state regulation.


Overview of Methodology


This Practice Guideline was developed using the RAND Corporation (RAND)/University of California, Los Angeles (UCLA) Appropriateness Method (RAM)—a process that combines scientific evidence and clinical knowledge to determine the appropriateness of a set of clinical procedures. The RAM is a deliberate approach encompassing review of existing guidelines, literature reviews, appropriateness ratings, necessity reviews, and document development. For this project, ASAM selected an independent committee to oversee guideline development, to participate in review of treatment scenarios, and to assist in writing. ASAM’s Quality Improvement Council, chaired by Margaret Jarvis, MD, oversaw the selection process for the independent development committee, referred to as the Guideline Committee.


The Guideline Committee comprised 10 experts and researchers from multiple disciplines, medical specialties, and subspecialties, including academic research, internal medicine, family medicine, addiction medicine, addiction psychiatry, general psychiatry, obstetrics/gynecology, pharmacology, and clinical neurobiology. Physicians with both allopathic and osteopathic training were represented in the Guideline Committee. The Guideline Committee was assisted by a technical team of researchers from the Treatment Research Institute (TRI) affiliated with the University of Pennsylvania, and worked under the guidance of Dr. Kyle Kampman who led the TRI team as Principal Investigator in implementing the RAM.


Summary of Recommendations


Part 1: Assessment and Diagnosis of Opioid Use Disorder


Assessment Recommendations




  • (1)

    First clinical priority should be given to identifying and making appropriate referral for any urgent or emergent medical or psychiatric problem(s), including drug-related impairment or overdose.


  • (2)

    Completion of the patient’s medical history should include screening for concomitant medical conditions, including infectious diseases (hepatitis, HIV, and tuberculosis [TB]), acute trauma, and pregnancy.


  • (3)

    A physical examination should be completed as a component of the comprehensive assessment process. The prescriber (the clinician authorizing the use of a medication for the treatment of opioid use disorder) may conduct this physical examination him/herself, or, in accordance with the ASAM Standards, ensure that a current physical examination is contained within the patient medical record before a patient is started on a new medication for the treatment of his/her addiction.


  • (4)

    Initial laboratory testing should include a complete blood count, liver function tests, and tests for hepatitis C and HIV. Testing for TB and sexually transmitted infections should also be considered. Hepatitis B vaccination should be offered, if appropriate.


  • (5)

    The assessment of women presents special considerations regarding their reproductive health. Women of childbearing age should be tested for pregnancy, and all women of childbearing potential and age should be queried regarding methods of contraception, given the increase in fertility that results from effective opioid use disorder treatment.


  • (6)

    Patients being evaluated for addiction involving opioid use, and/or for possible medication use in the treatment of opioid use disorder, should undergo (or have completed) an assessment of mental health status and possible psychiatric disorders (as outlined in the ASAM Standards).


  • (7)

    Opioid use is often co-occurring with other substance-related disorders. An evaluation of past and current substance use and a determination of the totality of substances that surround the addiction should be conducted.


  • (8)

    The use of marijuana, stimulants, or other addictive drugs should not be a reason to suspend opioid use disorder treatment. However, evidence demonstrates that patients who are actively using substances during opioid use disorder treatment have a poorer prognosis. The use of benzodiazepines and other sedative hypnotics may be a reason to suspend agonist treatment because of safety concerns related to respiratory depression.


  • (9)

    A tobacco use query and counseling on cessation of tobacco products and electronic nicotine delivery devices should be completed routinely for all patients, including those who present for evaluation and treatment of opioid use disorder.


  • (10)

    An assessment of social and environmental factors should be conducted (as outlined in the ASAM Standards) to identify facilitators and barriers to addiction treatment, and specifically to pharmacotherapy. Before a decision is made to initiate a course of pharmacotherapy for the patient with opioid use disorder, the patient should receive a multidimensional assessment in fidelity with the ASAM Criteria: Treatment Criteria for Addictive, Substance-Related, and Co-occurring Conditions (the “ASAM Criteria”). Addiction should be considered a bio-psycho-social-spiritual illness, for which the use of medication(s) is but only one component of overall treatment.



Diagnosis Recommendations




  • (1)

    Other clinicians may diagnose opioid use disorder, but confirmation of the diagnosis by the provider with prescribing authority, and who recommends medication use, must be obtained before pharmacotherapy for opioid use disorder commences.


  • (2)

    Opioid use disorder is primarily diagnosed on the basis of the history provided by the patient and a comprehensive assessment that includes a physical examination.


  • (3)

    Validated clinical scales that measure withdrawal symptoms, for example, the Objective Opioid Withdrawal Scale (OOWS), the Subjective Opioid Withdrawal Scale (SOWS), and the Clinical Opioid Withdrawal Scale (COWS), may be used to assist in the evaluation of patients with opioid use disorder.


  • (4)

    Urine drug testing during the comprehensive assessment process, and frequently during treatment, is recommended. The frequency of drug testing is determined by a number of factors including the stability of the patient, the type of treatment, and the treatment setting.



Part 2: Treatment Options




  • (1)

    The choice of available treatment options for addiction involving opioid use should be a shared decision between clinician and patient.


  • (2)

    Clinicians should consider the patient’s preferences, past treatment history, and treatment setting when deciding between the use of methadone, buprenorphine, and naltrexone in the treatment of addiction involving opioid use. The treatment setting described as level 1 treatment in the ASAM Criteria may be a general outpatient location such as a clinician’s practice site. The setting described as level 2 in the ASAM Criteria may be an intensive outpatient treatment or partial hospitalization program housed in a specialty addiction treatment facility, a community mental health center, or another setting. The ASAM Criteria describes level 3 or level 4 treatment, respectively, as a residential addiction treatment facility or hospital.


  • (3)

    The venue in which treatment is provided is as important as the specific medication selected. Opioid treatment programs (OTPs) offer daily supervised dosing of methadone, and increasingly of buprenorphine. In accordance with the Federal law (21 CFR §1306.07), office-based opioid treatment (OBOT), which provides medication on a prescribed weekly or monthly basis, is limited to buprenorphine. Naltrexone can be prescribed in any setting by any clinician with the authority to prescribe any medication. Clinicians should consider a patient’s psychosocial situation, co-occurring disorders, and risk of diversion when determining whether OTP or OBOT is most appropriate.


  • (4)

    OBOT may not be suitable for patients with active alcohol use disorder or sedative, hypnotic, or anxiolytic use disorder (or who are undergoing treatment for addiction involving the use of alcohol or other sedative drugs, including benzodiazepines or benzodiazepine receptor agonists). It may also be unsuitable for persons who are regularly using alcohol or other sedatives, but do not have addiction or a specific substance use disorder related to that class of drugs. The prescribing of benzodiazepines or other sedative-hypnotics should be used with extreme caution in patients who are prescribed methadone or buprenorphine for the treatment of an opioid use disorder.


  • (5)

    Methadone is recommended for patients who may benefit from daily dosing and supervision in an OTP, or for patients for whom buprenorphine for the treatment of opioid use disorder has been used unsuccessfully in an OTP or OBOT setting.


  • (6)

    Oral naltrexone for the treatment of opioid use disorder is often adversely affected by poor medication adherence. Clinicians should reserve its use for patients who would be able to comply with special techniques to enhance their adherence, for example, observed dosing. Extended-release injectable naltrexone reduces, but does not eliminate, issues with medication adherence.



Part 3: Treating Opioid Withdrawal




  • (1)

    Using medications for opioid withdrawal management is recommended over abrupt cessation of opioids. Abrupt cessation of opioids may lead to strong cravings, which can lead to continued use.


  • (2)

    Patients should be advised about risk of relapse and other safety concerns from using opioid withdrawal management as standalone treatment for opioid use disorder. Opioid withdrawal management on its own is not a treatment method.


  • (3)

    Assessment of a patient undergoing opioid withdrawal management should include a thorough medical history and physical examination, focusing on signs and symptoms associated with opioid withdrawal.


  • (4)

    Opioid withdrawal management in cases in which methadone is used to manage withdrawal symptoms must be done in an inpatient setting or in an OTP. For short-acting opioids, tapering schedules that decrease in daily doses of prescribed methadone should begin with doses between 20 and 30 mg per day, and should be completed within 6–10 days.


  • (5)

    Opioid withdrawal management in cases in which buprenorphine is used to manage withdrawal symptoms should not be initiated until 12–18 hours after the last dose of a short-acting agonist such as heroin or oxycodone, and 24–48 hours after the last dose of a long-acting agonist such as methadone. A dose of buprenorphine sufficient to suppress withdrawal symptoms is given (this can be 4–16 mg per day) and then the dose is tapered. The duration of the tapering schedule can be as brief as 3–5 days or as long as 30 days or more.


  • (6)

    The use of combinations of buprenorphine and low doses of oral naltrexone to manage withdrawal and facilitate the accelerated introduction of extended-release injectable naltrexone has shown promise. More research will be needed before this can be accepted as standard practice.


  • (7)

    The Guideline Committee recommends, based on consensus opinion, the inclusion of clonidine as a practice to support opioid withdrawal. Clonidine is not FDA-approved for the treatment of opioid withdrawal, but it has been used extensively off-label for this purpose. Clonidine may be used orally or transdermally at doses of 0.1–0.3 mg every 6–8 hours, with a maximum dose of 1.2 mg daily to assist in the management of opioid withdrawal symptoms. Its hypotensive effects often limit the amount that can be used. Clonidine can be combined with other non-narcotic medications targeting specific opioid withdrawal symptoms such as benzodiazepines for anxiety, loperamide for diarrhea, acetaminophen or nonsteroidal anti-inflammatory medications (NSAIDs) for pain, and ondansetron or other agents for nausea.


  • (8)

    Opioid withdrawal management using anesthesia UROD is not recommended due to high risk for adverse events or death. Naltrexone-facilitated opioid withdrawal management can be a safe and effective approach, but should be used only by clinicians experienced with this clinical method, and in cases in which anesthesia or conscious sedation are not being employed.



Part 4: Methadone




  • (1)

    Methadone is a treatment option recommended for patients who are physiologically dependent on opioids, able to give informed consent, and who have no specific contraindications for agonist treatment when it is prescribed in the context of an appropriate plan that includes psychosocial intervention.


  • (2)

    The recommended initial dose for methadone ranges from 10 to 30 mg, with reassessment in 3–4 hours, and a second dose not to exceed 10 mg on the first day if withdrawal symptoms are persisting.


  • (3)

    The usual daily dosage of methadone ranges from 60 to 120 mg. Some patients may respond to lower doses and some patients may need higher doses. Dosage increases in 5- to 10-mg increments applied no more frequently than every 7 days (depending on clinical response) are necessary to avoid over sedation, toxicity, or even iatrogenic overdose deaths.


  • (4)

    The administration of methadone should be monitored because unsupervised administration can lead to misuse and diversion. OTP regulations require monitored medication administration until the patient’s clinical response and behavior demonstrates that the prescribing of nonmonitored doses is appropriate.


  • (5)

    Psychosocial treatment, although sometimes minimally needed, should be implemented in conjunction with the use of methadone in the treatment of opioid use disorder.


  • (6)

    Methadone should be reinstituted immediately if relapse occurs, or when an assessment determines that the risk of relapse is high for patients who previously received methadone in the treatment of opioid use disorder, but who are no longer prescribed such treatment.


  • (7)

    Strategies directed at relapse prevention are an important part of comprehensive addiction treatment and should be included in any plan of care for a patient receiving active opioid treatment or ongoing monitoring of the status of their addictive disease.


  • (8)

    Switching from methadone to another medication for the treatment of opioid use disorder may be appropriate if the patient experiences intolerable side effects or is not successful in attaining or maintaining treatment goals through the use of methadone.


  • (9)

    Patients switching from methadone to buprenorphine in the treatment of opioid use disorder should be on low doses of methadone before switching medications. Patients on low doses of methadone (30–40 mg per day or less) generally tolerate transition to buprenorphine with minimal discomfort, whereas patients on higher doses of methadone may experience significant discomfort in switching medications.


  • (10)

    Patients switching from methadone to oral naltrexone or extended-release injectable naltrexone must be completely withdrawn from methadone and other opioids before they can receive naltrexone. The only exception would apply when an experienced clinician receives consent from the patient to embark on a plan of naltrexone-facilitated opioid withdrawal management.


  • (11)

    Patients who discontinue agonist therapy with methadone or buprenorphine and then resume opioid use should be made aware of the risks associated with opioid overdose, and especially the increased risk of death.



Part 5: Buprenorphine




  • (1)

    Opioid-dependent patients should wait until they are experiencing mild to moderate opioid withdrawal before taking the first dose of buprenorphine to reduce the risk of precipitated withdrawal. Generally, buprenorphine initiation should occur at least 6–12 hours after the last use of heroin or other short-acting opioids, or 24–72 hours after their last use of long-acting opioids such as methadone.


  • (2)

    Induction of buprenorphine should start with a dose of 2–4 mg. Dosages may be increased in increments of 2–4 mg.


  • (3)

    Clinicians should observe patients in their offices during induction. Emerging research, however, suggests that many patients need “not” be observed and that home buprenorphine induction may be considered. Home-based induction is recommended only if the patient or prescribing physician is experienced with the use of buprenorphine. This is based on the consensus opinion of the Guideline Committee.


  • (4)

    Buprenorphine doses after induction and titration should be, on average, at least 8 mg per day. However, if patients are continuing to use opioids, consideration should be given to increasing the dose by 4–8 mg (daily doses of 12–16 mg or higher). The FDA approves dosing to a limit of 24 mg per day, and there is limited evidence regarding the relative efficacy of higher doses. In addition, the use of higher doses may increase the risk of diversion.


  • (5)

    Psychosocial treatment should be implemented in conjunction with the use of buprenorphine in the treatment of opioid use disorder.


  • (6)

    Clinicians should take steps to reduce the chance of buprenorphine diversion. Recommended strategies include frequent office visits (weekly in early treatment), urine drug testing, including testing for buprenorphine and metabolites, and recall visits for pill counts.


  • (7)

    Patients should be tested frequently for buprenorphine, other substances, and prescription medications. Accessing Prescription Drug Monitoring Program (PDMP) data may be useful for monitoring.


  • (8)

    Patients should be seen frequently at the beginning of their treatment. Weekly visits (at least) are recommended until patients are determined to be stable. There is no recommended time limit for treatment.


  • (9)

    Buprenorphine taper and discontinuation is a slow process and close monitoring is recommended. Buprenorphine tapering is generally accomplished over several months. Patients should be encouraged to remain in treatment for ongoing monitoring past the point of discontinuation.


  • (10)

    When considering a switch from buprenorphine to naltrexone, 7–14 days should elapse between the last dose of buprenorphine and the start of naltrexone to ensure that the patient is not physically dependent on opioids before starting naltrexone.


  • (11)

    When considering a switch from buprenorphine to methadone, there is no required time delay because the addition of a full mu-opioid agonist to a partial agonist does not typically result in any type of adverse reaction.


  • (12)

    Patients who discontinue agonist therapy and resume opioid use should be made aware of the risks associated with an opioid overdose, and especially the increased risk of death.



Part 6: Naltrexone




  • (1)

    Naltrexone is a recommended treatment in preventing relapse in opioid use disorder. Oral formula naltrexone may be considered for patients in whom adherence can be supervised or enforced. Extended-release injectable naltrexone may be more suitable for patients who have issues with adherence.


  • (2)

    Oral naltrexone should be taken daily in 50-mg doses, or three times weekly in two 100-mg doses followed by one 150-mg dose.


  • (3)

    Extended-release injectable naltrexone should be administered every 4 weeks by deep IM injection in the gluteal muscle at a set dosage of 380 mg per injection.


  • (4)

    Psychosocial treatment is recommended in conjunction with treatment with naltrexone. The efficacy of naltrexone use in conjunction with psychosocial treatment has been established, whereas the efficacy of extended release injectable naltrexone without psychosocial treatment “has not” been established.


  • (5)

    There is no recommended length of treatment with oral naltrexone or extended-release injectable naltrexone. Duration depends on clinical judgment and the patient’s individual circumstances. Because there is no physical dependence associated with naltrexone, it can be stopped abruptly without withdrawal symptoms.


  • (6)

    Switching from naltrexone to methadone or buprenorphine should be planned, considered, and monitored. Switching from an antagonist such as naltrexone to a full agonist (methadone) or a partial agonist (buprenorphine) is generally less complicated than switching from a full or partial agonist to an antagonist because there is no physical dependence associated with antagonist treatment and thus no possibility of precipitated withdrawal. Patients being switched from naltrexone to buprenorphine or methadone will not have physical dependence on opioids and thus the initial doses of methadone or buprenorphine used should be low. Patients should not be switched until a significant amount of the naltrexone is no longer in their system, about 1 day for oral naltrexone or 30 days for extended-release injectable naltrexone.


  • (7)

    Patients who discontinue antagonist therapy and resume opioid use should be made aware of the increased risks associated with an opioid overdose, and especially the increased risk of death.



Part 7: Psychosocial Treatment in Conjunction With Medications for the Treatment of Opioid Use Disorder




  • (1)

    Psychosocial treatment is recommended in conjunction with any pharmacological treatment of opioid use disorder. At a minimum, psychosocial treatment should include the following: psychosocial needs assessment, supportive counseling, links to existing family supports, and referrals to community services.


  • (2)

    Treatment planning should include collaboration with qualified behavioral healthcare providers to determine the optimal type and intensity of psychosocial treatment and for renegotiation of the treatment plan for circumstances in which patients do not adhere to recommended plans for, or referrals to, psychosocial treatment.


  • (3)

    Psychosocial treatment is generally recommended for patients who are receiving opioid agonist treatment (methadone or buprenorphine).


  • (4)

    Psychosocial treatment is recommended in the treatment of pregnant women with opioid use disorder.


  • (5)

    Counseling and testing for HIV should be provided in accordance with state law. Tests for hepatitis B and C and liver function are also suggested. Hepatitis A and B vaccination is recommended for those whose hepatitis serology is negative.


  • (6)

    Urine drug testing may be used to detect or confirm suspected opioid and other drug use with informed consent from the mother, realizing that there may be adverse legal and social consequences of her use. State laws differ on reporting substance use during pregnancy. Laws that penalize women for use and for obtaining treatment serve to prevent women from obtaining prenatal care and worsen outcomes.


  • (7)

    Pregnant women who are physically dependent on opioids should receive treatment using methadone or buprenorphine monoproduct rather than withdrawal management or abstinence.


  • (8)

    Care for pregnant women with opioid use disorder should be comanaged by an obstetrician and an addiction specialist physician. Release of information forms need to be completed to ensure communication among healthcare providers.


  • (9)

    Treatment with methadone should be initiated as early as possible during pregnancy.


  • (10)

    Hospitalization during initiation of methadone and treatment with buprenorphine may be advisable due to the potential for adverse events, especially in the third trimester.


  • (11)

    In an inpatient setting, methadone should be initiated at a dose range of 20–30 mg. Incremental doses of 5–10 mg are given every 3–6 hours, as needed, to treat withdrawal symptoms.


  • (12)

    After induction, clinicians should increase the methadone dose in 5- to 10-mg increments per week. The goal is to maintain the lowest dose that controls withdrawal symptoms and minimizes the desire to use additional opioids.


  • (13)

    Twice daily dosing is more effective and has fewer side effects than single dosing but may not be practical because methadone is typically dispensed in an outpatient clinic.


  • (14)

    Clinicians should be aware that the pharmacokinetics of methadone are affected by pregnancy. With advancing gestational age, plasma levels of methadone progressively decrease and clearance increases. Increased or split doses may be needed as pregnancy progresses. After child birth, doses may need to be adjusted.


  • (15)

    Buprenorphine monoproduct is a reasonable and recommended alternative to methadone for pregnant women. Whereas there is evidence of safety, there is insufficient evidence to recommend the combination buprenorphine/naloxone formulation.


  • (16)

    If a woman becomes pregnant while she is receiving naltrexone, it is appropriate to discontinue the medication if the patient and doctor agree that the risk of relapse is low. If the patient is highly concerned about relapse and wishes to continue naltrexone, she should be informed about the risks of staying on naltrexone and provide her consent for ongoing treatment. If the patient wishes to discontinue naltrexone, but then reports relapse to opioid use, it may be appropriate to consider treatment with methadone or treatment with buprenorphine.


  • (17)

    Naloxone is not recommended for use in pregnant women with opioid use disorder except in situations of life-threatening overdose.


  • (18)

    Mothers receiving methadone and buprenorphine monoproduct for the treatment of opioid use disorders should be encouraged to breastfeed.



Part 9: Special Populations: Individuals With Pain




  • (1)

    For all patients with pain, it is important that the correct diagnosis be made and that a target suitable for treatment is identified.


  • (2)

    If pharmacological treatment is considered, non-narcotic medications such as acetaminophen and NSAIDs should be tried first.


  • (3)

    Opioid agonists (methadone or buprenorphine) should be considered for patients with active opioid use disorder who are not under treatment.


  • (4)

    Pharmacotherapy in conjunction with psychosocial treatment should be considered for patients with pain who have opioid use disorder.


  • (5)

    Patients on methadone for the treatment of opioid use disorder will require doses of opioids in addition to their regular daily dose of methadone to manage acute pain.


  • (6)

    Patients on methadone for the treatment of opioid use disorder and who are admitted for surgery may require additional short-acting opioid pain relievers. The dose of pain relievers prescribed may be higher due to tolerance.


  • (7)

    Temporarily increasing buprenorphine dosing may be effective for mild acute pain.


  • (8)

    For severe acute pain, discontinuing buprenorphine and commencing on a high-potency opioid (such as fentanyl) is advisable. Patients should be monitored closely and additional interventions such as regional anesthesia should also be considered.


  • (9)

    The decision to discontinue buprenorphine before an elective surgery should be made in consultation with the attending surgeon and anesthesiologist. If it is decided that buprenorphine should be discontinued before surgery, this should occur 24–36 hours in advance of surgery and restarted postoperatively when the need for full opioid agonist analgesia has passed.


  • (10)

    Patients on naltrexone will not respond to opioid analgesics in the usual manner. Therefore, it is recommended that mild pain be treated with NSAIDs, and moderate to severe pain be treated with ketorolac on a short-term basis.


  • (11)

    Oral naltrexone should be discontinued 72 hours before surgery and extended-release injectable naltrexone should be discontinued 30 days before an anticipated surgery.



Part 10: Special Populations: Adolescents




  • (1)

    Clinicians should consider treating adolescents who have opioid use disorder using the full range of treatment options, including pharmacotherapy.


  • (2)

    Opioid agonists (methadone and buprenorphine) and antagonists (naltrexone) may be considered for treatment of opioid use disorder in adolescents. Age is a consideration in treatment, and Federal laws and US FDA approvals need to be considered for patients under age 18.


  • (3)

    Psychosocial treatment is recommended in the treatment of adolescents with opioid use disorder.


  • (4)

    Concurrent practices to reduce infection (e.g., sexual risk reduction interventions) are recommended as components of comprehensive treatment for the prevention of sexually transmitted infections and blood-borne viruses.


  • (5)

    Adolescents may benefit from treatment in specialized treatment facilities that provide multidimensional services.



Part 11: Special Populations: Individuals With Co-occurring Psychiatric Disorders




  • (1)

    A comprehensive assessment including determination of mental health status should evaluate whether the patient is stable. Patients with suicidal or homicidal ideation should be referred immediately for treatment and possibly hospitalization.


  • (2)

    Management of patients at risk for suicide should include reducing immediate risk; managing underlying factors associated with suicidal intent; and monitoring and follow-up.


  • (3)

    All patients with psychiatric disorders should be asked about suicidal ideation and behavior. Patients with a history of suicidal ideation or attempts should have opioid use disorder, and psychiatric medication use, monitored.


  • (4)

    Assessment for psychiatric disorder should occur at the onset of agonist or antagonist treatment. Reassessment using a detailed mental status examination should occur after stabilization with methadone, buprenorphine, or naltrexone.


  • (5)

    Pharmacotherapy in conjunction with psychosocial treatment should be considered for patients with opioid use disorder and a co-occurring psychiatric disorder.


  • (6)

    Clinicians should be aware of potential interactions between medications used to treat co-occurring psychiatric conditions and opioid use disorder.


  • (7)

    Assertive community treatment should be considered for patients with co-occurring schizophrenia and opioid use disorder who have a recent history of, or are at risk of, repeated hospitalization or homelessness.



Part 12: Special Populations: Individuals in the Criminal Justice System




  • (1)

    Pharmacotherapy for the continued treatment of opioid use disorders, or the initiation of pharmacotherapy, has been shown to be effective and is recommended for prisoners and parolees regardless of the length of their sentenced term.


  • (2)

    Individuals with opioid use disorder who are within the criminal justice system should be treated with some type of pharmacotherapy in addition to psychosocial treatment.


  • (3)

    Opioid agonists (methadone and buprenorphine) and antagonists (naltrexone) may be considered for treatment. There is insufficient evidence to recommend any one treatment as superior to another for prisoners or parolees.


  • (4)

    Pharmacotherapy should be initiated a minimum of 30 days before release from prison.



Part 13: Naloxone for the Treatment of Opioid Overdose




  • (1)

    Naloxone should be given in case of opioid overdose.


  • (2)

    Naloxone can and should be administered to pregnant women in cases of overdose to save the mother’s life.


  • (3)

    The Guideline Committee, based on consensus opinion, recommends that patients who are being treated for opioid use disorder and their family members/significant others be given prescriptions for naloxone. Patients and family members/significant others should be trained in the use of naloxone in overdose.


  • (4)

    The Guideline Committee, based on consensus opinion, recommends that first responders such as emergency medical services personnel, police officers, and firefighters be trained in and authorized to administer naloxone.



Abbreviations and Acronyms





  • AA Alcoholics Anonymous



  • ACT assertive community treatment



  • AIDS acquired immunodeficiency syndrome



  • ASAM American Society of Addiction Medicine



  • CBT cognitive behavioral therapy



  • CDC Centers for Disease Control and Prevention



  • COWS Clinical Opioid Withdrawal Scale



  • DATA 2000 Drug Addiction Treatment Act of 2000



  • DEA Drug Enforcement Administration



  • DSM-III Diagnostic and Statistical Manual of Mental Disorders, Third Edition



  • DSM-IV Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition



  • DSM-5 Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition



  • ECG electrocardiogram



  • EMS emergency medical services



  • FDA US Food and Drug Administration



  • HBV hepatitis B virus



  • HCV hepatitis C virus



  • HIV human immunodeficiency virus



  • IDU injection drug use



  • IM intramuscular



  • IV intravenous



  • NA Narcotics Anonymous



  • NAS neonatal abstinence syndrome



  • NSAIDs nonsteroidal anti-inflammatory drugs



  • NSDUH National Survey on Drug Use and Health



  • OBOT office-based opioid treatment



  • OOWS Objective Opioid Withdrawal Scale



  • OTP opioid treatment program



  • PMDP prescription drug monitoring program



  • RCT randomized clinical trial



  • RAM RAND/UCLA Appropriateness Method



  • SAMHSA Substance Abuse and Mental Health Services Administration



  • SMART self-management and recovery therapy



  • SOWS Subjective Opioid Withdrawal Scale



  • TB tuberculosis



  • UROD ultra-rapid opioid detoxification



National Practice Guideline Glossary





  • Abstinence: Intentional and consistent restraint from the pathological pursuit of reward and/or relief that involves the use of substances and other behaviors. These behaviors may involve, but are not necessarily limited to, gambling, video gaming, spending, compulsive eating, compulsive exercise, or compulsive sexual behaviors. 111



  • Abuse: This term is not recommended for use in clinical or research contexts. Harmful use of a specific psychoactive substance. When used to mean “substance abuse,” this term also applies to one category of psychoactive substance-related disorders in previous editions of the Diagnostic and Statistical Manual of Mental Disorders (DSM). While recognizing that “abuse” is part of past diagnostic terminology, ASAM recommends that an alternative term be found for this purpose because of the pejorative connotations of the word “abuse.” 111



  • Addiction: Addiction is a primary, chronic disease of brain reward, motivation, memory, and related circuitry. Dysfunction in these circuits leads to characteristic biological, psychological, social, and spiritual manifestations. This is reflected in an individual pathologically pursuing reward and/or relief by substance use and other behaviors.


    Addiction is characterized by inability to consistently abstain, impairment in behavioral control, cravings, diminished recognition of significant problems with one’s behaviors and interpersonal relationships, and a dysfunctional emotional response. Like other chronic diseases, addiction often involves cycles of relapse and remission. Without treatment or engagement in recovery activities, addiction is progressive and can result in disability or premature death. 111



  • Addiction specialist physician: Addiction specialist physicians include addiction medicine physicians and addiction psychiatrists who hold either a board certification in addiction medicine from the American Board of Addiction Medicine, a subspecialty board certification in addiction psychiatry from the American Board of Psychiatry and Neurology, a subspecialty board certification in addiction medicine from the American Osteopathic Association, or certification in addiction medicine from the American Society of Addiction Medicine. 118



  • Adherence (see also Compliance): To “adhere” is “to cling, cleave (to be steadfast, hold fast), to stick fast” (Webster’s Dictionary). Adherence is a term that health professionals have been using increasingly to replace the term “compliance.” Both terms have been used, sometimes interchangeably, to refer to how closely patients cooperate with, follow, and take personal responsibility for the implementation of their treatment plans. The terms are often used with the more narrow sense of how well patients accomplish the goal of persistently taking medications, and also refer more broadly to all components of treatment. Assessment of patients’ efforts to accomplish the goals of a treatment plan is essential to treatment success. These efforts occur along a complex spectrum from independent proactive commitment, to mentored collaboration, to passive cooperation, to reluctant partial agreement, to active resistance, and to full refusal. Attempts to understand factors that promote or inhibit adherence/compliance must take into account behaviors, attitudes, willingness, and varying degrees of capacity and autonomy. The term “adherence” emphasizes the patient’s collaboration and participation in treatment. It contributes to a greater focus on motivational enhancement approaches that engage and empower patients. 111



  • Adolescence: The American Academy of Pediatrics categorizes adolescence as the totality of three developmental stages—puberty to adulthood—which occur generally between 11 and 21 years of age. 71



  • Agonist medication: See Opioid agonist medication.



  • Antagonist medication: See Opioid antigonist medication.



  • ASAM Criteria dimensions: The ASAM Patient Placement Criteria use six dimensions to create a holistic biopsychosocial assessment of an individual to be used for service planning and treatment. Dimension one is acute intoxication or withdrawal potential. Dimension two is biomedical conditions and conditions. Dimension three is emotional, behavioral, or cognitive conditions or complications. Dimension four is readiness for change. Dimension five is continued use or continued problem potential. Dimension six is recovery/living environment. 111



  • Assertive community treatment: An evidence-based, outreach-oriented, service delivery model for people with severe and persistent mental illnesses that uses a team-based model to provide comprehensive and flexible treatment. 13



  • Clinician: A health professional, such as a physician, psychiatrist, psychologist, or nurse, involved in clinical practice, as distinguished from one specializing in research. 111



  • Cognitive behavioral therapy : An evidence-based psychosocial intervention that seeks to modify harmful beliefs and maladaptive behaviors, and help patients recognize, avoid, and cope with the situations in which they are most likely to misuse drugs. 149



  • Co-occurring disorders: Concurrent substance use and mental disorders. Other terms used to describe co-occurring disorders include “dual diagnosis,” “dual disorders,” “mentally ill chemically addicted” (MICA), “chemically addicted mentally ill” (CAMI), “mentally ill substance abusers” (MISA), “mentally ill chemically dependent” (MICD), “concurrent disorders,” “coexisting disorders,” “comorbid disorders,” and “individuals with co-occurring psychiatric and substance symptomatology” (ICOPSS). Use of the term carries no implication as to which disorder is primary and which secondary, which disorder occurred first, or whether one disorder caused the other. 111



  • Compliance (see also Adherence): “To comply” is “to act in accordance with another’s wishes, or with rules and regulations” (Webster’s Dictionary). The term “compliance” is falling into disuse because patient engagement and responsibility to change is a goal beyond passive compliance. Given the importance of shared decision-making to improve collaboration and outcomes, patients are encouraged to actively participate in treatment decisions and take responsibility for their treatment, rather than to passively comply. 111



  • Concomitant conditions: Medical conditions (e.g., HIV, cardiovascular disease) and/or psychiatric conditions (e.g., depression, schizophrenia) that occur along with a substance use disorder. 120



  • Contingency management: An evidence-based psychosocial intervention in which patients are given tangible rewards to reinforce positive behaviors such as abstinence. Also referred to as motivational incentives. 149



  • Dependence: Used in three different ways: physical dependence is a state of adaptation that is manifested by a drug class-specific withdrawal syndrome that can be produced by abrupt cessation, rapid dose reduction, decreasing blood level of the drug, and/or administration of an antagonist; psychological dependence is a subjective sense of need for a specific psychoactive substance, either for its positive effects or to avoid negative effects associated with its abstinence; and one category of psychoactive substance use disorder in previous editions of the DSM, but not in DSM-5. 111



  • Detoxification: Usually used to refer to a process of withdrawing a person from a specific psychoactive substance in a safe and effective manner. The term actually encompasses safe management of intoxication states (more literally, “detoxification”) and of withdrawal states. In this document, this term has been replaced by the term Withdrawal Management. 111



  • Failure (as in treatment failure): This term is not recommended for use in clinical or research contexts. Lack of progress and/or regression at any given level of care. Such a situation warrants a reassessment of the treatment plan, with modification of the treatment approach. Such situations may require changes in the treatment plan at the same level of care or transfer to a different (more or less intensive) level of care to achieve a better therapeutic response and outcome. Sometimes used to describe relapse after a single treatment episode—an inappropriate construct in describing a chronic disease or disorder. The use of “treatment failure” is therefore not a recommended concept or term to be used. 111



  • Harm reduction: A treatment and prevention approach that encompasses individual and public health needs, aiming to decrease the health and socioeconomic costs and consequences of addiction-related problems, especially medical complications and transmission of infectious diseases, without necessarily requiring abstinence. Abstinence-based treatment approaches are themselves a part of comprehensive harm reduction strategies. A range of recovery activities may be included in every harm reduction strategy. 111



  • Induction (office and home): The phase of opioid treatment during which maintenance medication dosage levels are adjusted until a patient attains stabilization. Buprenorphine induction may take place in an office-based setting or home-based setting. Methadone induction must take place in an opioid treatment program (OTP). 152



  • Illicit opioid (nonmedical drug use): Use of an illicit drug or the use of a prescribed medicine for reasons other than the reasons intended by the prescriber, for example, to produce positive reward or negative reward. Nonmedical use of prescription drugs often includes use of a drug in higher doses than authorized by the prescriber or through a different route of administration than intended by the prescriber, and for a purpose other than the indication intended by the prescriber (e.g. the use of methylphenidate prescribed for attention-deficit/hyperactivity disorder [ADHD] to produce euphoria rather than to reduce symptoms or dysfunction from ADHD). 6



  • Maintenance treatment(s): Pharmacotherapy on a consistent schedule for persons with addiction, usually with an agonist or partial agonist, which militates against the pathological pursuit of reward and/or relief and allows remission of overt addiction-related problems.


    Maintenance treatments of addiction are associated with the development of a pharmacological steady state in which receptors for addictive substances are occupied, resulting in relative or complete blockade of central nervous system receptors such that addictive substances are no longer sought for reward and/or relief. Maintenance treatments of addiction are also designed to militate against the risk of overdose. Depending on the circumstances of a given case, a care plan including maintenance treatments can be time-limited or can remain in place lifelong. Integration of pharmacotherapy via maintenance treatments with psychosocial treatment generally is associated with the best clinical results. Maintenance treatments can be part of an individual’s treatment plan in abstinence-based recovery activities or can be a part of harm reduction strategies. 111



  • Moderation management: Moderation management (MM) is a behavioral change program and national support group network for people concerned about their drinking and who desire to make positive lifestyle changes. MM empowers individuals to accept personal responsibility for choosing and maintaining their own path, whether moderation or abstinence. MM promotes early self-recognition of risky drinking behavior, when moderate drinking is a more easily achievable goal. 116



  • Motivational interviewing:


  • (1)

    Layperson’s definition: A collaborative conversation style for strengthening a person’s own motivation and commitment to change.


  • (2)

    Practitioner’s definition: A person-centered counseling style for addressing the common problem of ambivalence about change.


  • (3)

    Technical definition: A collaborative, goal-oriented style of communication with particular attention to the language of change. It is designed to strengthen personal motivation for and commitment to a specific goal by eliciting and exploring the person’s own reasons for change within an atmosphere of acceptance and compassion. 111



  • Naloxone challenge: Naloxone is a short-acting opioid antagonist. Naloxone challenge is a test in which naloxone is administered to patients to evaluate their level of opioid dependence before the commencement of opioid pharmacotherapy. 152,170



  • Naltrexone-facilitated opioid withdrawal management: This is a method of withdrawal management. It involves the use of a single dose of buprenorphine combined with multiple small doses of naltrexone over a several day period to manage withdrawal and facilitate the initiation of treatment with naltrexone. 136



  • Narcotic drugs: Legally defined by the Controlled Substances Act in the United States since its enactment in 1970. The term “narcotic” is broad and can include drugs produced directly or indirectly by extraction from substances of vegetable origin, or independently by means of chemical synthesis, or by a combination of extraction and chemical synthesis. The main compounds defined as narcotics in the United States include opium opiates, derivatives of opium and opiates, including their isomers, esters, ethers, salts, and salts of isomers, esters, ethers (but not the isoquinoline alkaloids of opium), poppy straw and concentrate of poppy straw, coca leaves, cocaine, its salts, optical and geometric isomers, and salts of isomers and ecgonine, its derivatives, their salts, isomers, and salts of isomers. Any compound, mixture, or preparation which contains any quantity of any of the substances referred to above. 54



  • Neuroadaption: See “Tolerance” for the definition.



  • Office-based opioid treatment (OBOT): Physicians in private practices or a number of types of public sector clinics can be authorized to prescribe outpatient supplies of the partial opioid agonist buprenorphine. There is no regulation per se of the clinic site itself, but of the individual physician who prescribes buprenorphine. 111



  • Opiate: One of a group of alkaloids derived from the opium poppy ( Papaver somniferum ), with the ability to induce analgesia, euphoria, and, in higher doses, stupor, coma, and respiratory depression. The term excludes synthetic opioids. 170



  • Opioid: A current term for any psychoactive chemical that resembles morphine in pharmacological effects, including opiates and synthetic/semisynthetic agents that exert their effects by binding to highly selective receptors in the brain where morphine and endogenous opioids affect their actions. 6



  • Opioid agonist medication: Opioid agonist medications pharmacologically occupy opioid receptors in the body. They thereby relieve withdrawal symptoms and reduce or extinguish cravings for opioids. 111



  • Opioid antagonist medication: Opioid antagonist medications pharmacologically occupy opioid receptors in the body, but do not activate the receptors. This effectively blocks the receptor, preventing the brain from responding to opioids. The result is that further use of opioids does not produce euphoria or intoxication. 111



  • Opioid intoxication: A condition that follows the administration of opioids, resulting in disturbances in the level of consciousness, cognition, perception, judgment, affect, behavior, or other psychophysiological functions and responses. These disturbances are related to the acute pharmacological effects of, and learned responses to, opioids. With time, these disturbances resolve, resulting in complete recovery, except when tissue damage or other complications have arisen. Intoxication depends on the type and dose of opioid, and is influenced by factors such as an individual’s level of tolerance. Individuals often take drugs in the quantity required to achieve a desired degree of intoxication. Behavior resulting from a given level of intoxication is strongly influenced by cultural and personal expectations about the effects of the drug. According to the International Classifications of Diseases, Tenth Revision (ICD-10), acute intoxication is the term used for intoxication of clinical significance (F11.0). Complications may include trauma, inhalation of vomitus, delirium, coma, and convulsions, depending on the substance and method of administration. 170



  • Opioid treatment program (OTP): A program certified by the United States, Substance Abuse and Mental Health Services Administration (SAMHSA), usually comprising a facility, staff, administration, patients, and services, that engages in supervised assessment and treatment, using methadone, buprenorphine, L-alpha acetyl methadol, or naltrexone, of individuals who are addicted to opioids. An OTP can exist in a number of settings including, but not limited to, intensive outpatient, residential, and hospital settings. Services may include medically supervised withdrawal and/or maintenance treatment, along with various levels of medical, psychiatric, psychosocial, and other types of supportive care. 152



  • Opioid treatment services (OTS): An umbrella term that encompasses a variety of pharmacological and nonpharmacological treatment modalities. This term broadens understanding of opioid treatments to include all medications used to treat opioid use disorders and the psychosocial treatment that is offered concurrently with these pharmacotherapies. Pharmacological agents include opioid agonist medications such as methadone and buprenorphine, and opioid antagonist medications such as naltrexone. 111



  • Opioid use disorder: A substance use disorder involving opioids. See Substance Use Disorder.



  • Opioid withdrawal syndrome: Over time, morphine and its analogs induce tolerance and neuroadaptive changes that are responsible for rebound hyperexcitability when the drug is withdrawn. The withdrawal syndrome includes craving, anxiety, dysphoria, yawning, sweating, piloerection (gooseflesh), lacrimation (excessive tear formation), rhinorrhea (running nose), insomnia, nausea or vomiting, diarrhea, cramps, muscle aches, and fever. With short-acting drugs, such as morphine or heroin, withdrawal symptoms may appear within 8–12 hours of the last dose of the drug, reach a peak at 48–72 hours, and clear after 7–10 days. With longer-acting drugs, such as methadone, onset of withdrawal symptoms may not occur until 1–3 days after the last dose; symptoms peak between the third and eighth day and may persist for several weeks, but are generally milder than those that follow morphine or heroin withdrawal after equivalent doses. 170



  • Overdose: The inadvertent or deliberate consumption of a dose much larger than that either habitually used by the individual or ordinarily used for treatment of an illness, and likely to result in a serious toxic reaction or death. 111



  • Patient: As used in this document, an individual receiving alcohol, tobacco, and/or other drug or addictive disorder treatment. The terms “client” and “patient” sometimes are used interchangeably, although staff in nonmedical settings more commonly refer to “clients.” 111



  • Physical dependence: State of physical adaptation that is manifested by a drug class-specific withdrawal syndrome that can be produced by abrupt cessation, rapid dose reduction and/or decreasing blood level of a substance and/or administration of an antagonist. 152



  • Psychosocial treatment: Any nonpharmacological intervention carried out in a therapeutic context at an individual, family, or group level. Psychosocial interventions may include structured, professionally administered interventions (e.g., cognitive behavior therapy or insight-oriented psychotherapy) or nonprofessional interventions (e.g., self-help groups and nonpharmacological interventions from traditional healers). 13



  • Precipitated withdrawal: A condition that occurs when an opioid agonist is displaced from the opioid receptors by an antagonist. It is also possible for a partial agonist to precipitate withdrawal. 170



  • Recovery: A process of sustained action that addresses the biological, psychological, social, and spiritual disturbances inherent in addiction. This effort is in the direction of a consistent pursuit of abstinence, addressing impairment in behavioral control, dealing with cravings, recognizing problems in one’s behaviors and interpersonal relationships, and dealing more effectively with emotional responses. Recovery actions lead to reversal of negative, self-defeating internal processes and behaviors, allowing healing of relationships with self and others. The concepts of humility, acceptance, and surrender are useful in this process. (Note: ASAM continues to explore, as an evolving process, improved ways to define Recovery.) 111



  • Relapse: A process in which an individual who has established abstinence or sobriety experiences recurrence of signs and symptoms of active addiction, often including resumption of the pathological pursuit of reward and/or relief through the use of substances and other behaviors. When in relapse, there is often disengagement from recovery activities. Relapse can be triggered by exposure to rewarding substances and behaviors, by exposure to environmental cues to use, and by exposure to emotional stressors that trigger heightened activity in brain stress circuits. The event of using or acting out is the latter part of the process, which can be prevented by early intervention. 111



  • Sedative, hypnotic, or anxiolytics: This class of substances includes all prescription sleeping medications and virtually all prescription antianxiety medications. Nonbenzodiazepine antianxiety medications, such as buspirone and gepirone, are not included in this class because they are not associated with significant misuse. 160



  • Sobriety: A state of sustained abstinence with a clear commitment to and active seeking of balance in the biological, psychological, social, and spiritual aspects of an individual’s health and wellness that were previously compromised by active addiction. 111



  • Spontaneous withdrawal: A condition that occurs when an individual who is physically dependent on an opioid agonist suddenly discontinues or markedly decreases opioid use. 158



  • Stabilization: Includes the medical and psychosocial processes of assisting the patient through acute intoxication and withdrawal to the attainment of a medically stable, fully supported, substance-free state. This often is done with the assistance of medications, although in some approaches to detoxification, no medication is used. 152



  • Substance use disorder: Substance use disorder is marked by a cluster of cognitive, behavioral, and physiological symptoms indicating that the individual continues to use alcohol, tobacco, and/or other drugs despite significant related problems. Diagnostic criteria are given in the DSM-5. Substance use disorder is the new nomenclature for what was included as substance dependence and substance abuse in the DSM-IV. 6



  • Tolerance: A decrease in response to a drug dose that occurs with continued use. If an individual is tolerant to a drug, increased doses are required to achieve the effects originally produced by lower doses. Both physiological and psychosocial factors may contribute to the development of tolerance. Physiological factors include metabolic and functional tolerance. In metabolic tolerance, the body can eliminate the substance more readily, because the substance is metabolized at an increased rate. In functional tolerance, the central nervous system is less sensitive to the substance. An example of a psychosocial factor contributing to tolerance is behavioral tolerance, when learning or altered environmental constraints change the effect of the drug. Acute tolerance refers to rapid, temporary accommodation to the effect of a substance after a single dose. Reverse tolerance, also known as sensitization, refers to a condition in which the response to a substance increased with repeated use. Tolerance is one of the criteria of the dependence syndrome. 170



  • Withdrawal management: Withdrawal management describes services to assist a patient’s withdrawal. The liver detoxifies, but clinicians manage withdrawal. 118





Introduction


Purpose


The American Society of Addiction Medicine (ASAM) developed the National Practice Guideline for the Use of Medications in the Treatment of Addiction Involving Opioid Use (the “Practice Guideline”) to provide information on evidence-based treatment of opioid use disorder. This guideline is intended to assist clinicians in the decision-making process for prescribing pharmacotherapies and psychosocial treatments to patients with opioid use disorder.


Specifically, the Practice Guideline helps in the following:



  • (1)

    Identifies current practices and outstanding questions regarding the safe and effective use of medications for the treatment of opioid use disorder.


  • (2)

    Uses a methodology that integrates evidence-based practices and expert clinical judgment to develop recommendations on best practices in opioid use disorder treatment.


  • (3)

    Presents best practices in a cohesive document for clinicians’ use to improve the effectiveness of opioid use disorder treatment.



Background on Opioid Use Disorder


Opioid use disorder is a chronic, relapsing disease, which has significant economic, personal, and public health consequences. Many readers of this Practice Guideline may recognize the term “opioid use disorder” as it is used in the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition (DSM-5) developed by the American Psychiatric Association; others may be more familiar with the term “opioid dependence,” as used in previous editions of the DSM.


The ASAM defines addiction as “a primary, chronic disease of brain reward, motivation, memory, and related circuitry,” with a “dysfunction in these circuits” being reflected in “an individual pathologically pursuing reward and/or relief of withdrawal symptoms by substance use and other behaviors.” In this context, the preferred term by ASAM for this serious bio-psycho-social-spiritual illness would be “addiction involving opioid use.” ASAM views addiction as a fundamental neurological disorder of “brain reward, motivation, memory, and related circuitry,” and recognizes that there are unifying features in all cases of addiction, including substance-related addiction and nonsubstance-related addiction. It is clear that a variety of substances commonly associated with addiction work on specific receptors in the nervous system and on specific neurotransmitter systems. Specific pharmacological agents used in the treatment of addiction exert their effects via their actions on specific receptors. Hence, the medications used in the treatment of addiction have specific efficacy based on their own molecular structure and the particular neurotransmitters affected by that medication. Medications developed for the treatment of addiction involving opioid use may have benefits in the treatment of addiction involving an individual’s use of other substances. For instance, naltrexone (US Food and Drug Administration [FDA]), for the treatment of opioid dependence using DSM, Fourth Edition (DSM-IV) terminology, is also FDA-approved for the treatment of alcohol dependence, as per the DSM-IV guidelines.


The ASAM recognizes that research is yet to be done to confirm the specificity of its conceptualization of addiction as a medical and a psychiatric illness (note: the International Classification of Diseases-10 [ICD-10], and the American Medical Association in various policy and position statements recognize addiction as both a medical and a psychiatric disorder). ASAM encourages clinicians, researchers, educators, and policymakers to use the term “addiction” regardless of whether the patient’s condition at a given point in its natural history appears to more prominently involve opioid use or alcohol use, nicotine use, or engagement in addictive behaviors such as gambling. Given the widespread North American application of the DSM’s categorization of disorders, this Practice Guideline will for the sake of brevity and convention, use the term “opioid use disorder.”


Epidemiology


According to the 2013 National Survey on Drug Use and Health (NSDUH), 146 4.5 million individuals were current nonmedical users of prescription opioids (past month) and 1.9 million individuals met DSM-IV criteria for abuse or dependence of prescription opioids. In addition, the NSDUH reported that 289,000 people were current (past month) users of heroin and 517,000 met DSM IV criteria for abuse or dependence in 2013. The rate of prescription opioid use for nonmedical purposes was 1.7% in persons 12 years and older. However, the rate of prescription opioid use among youth aged 12–17 declined from 3.2% in 2002 and 2003 to 1.7% in 2013.


It is important to note that nonmedical use of prescription opioids has been shown to be associated with the initiation of heroin use. In a study pooling data from the NSDUH from 2002 to 2012, the incidence of heroin use was 19 times greater among individuals who reported prior nonmedical use of prescription opioids compared to individuals who did not report prior nonmedical prescription opioid use. 117


Mortality and Morbidity


Opioid use is associated with increased mortality. The leading causes of death in people using opioids for nonmedical purposes are overdose and trauma. 50 The number of unintentional overdose deaths from prescription opioids has more than quadrupled since 1999. 127


Opioid use increases the risk of exposure to HIV, viral hepatitis, and other infectious agents through contact with infected blood or body fluids (e.g., semen) that results from sharing syringes and injection paraphernalia, or through unprotected sexual contact. Similarly, it increases the risk of contracting infectious diseases such as HIV/AIDS and hepatitis because people under the influence of drugs may engage in risky behaviors that can expose them to these diseases. 50


It is notable that injection drug use (IDU) is the highest-risk behavior for acquiring hepatitis C virus (HCV) infection and continues to drive this epidemic. Of the 17,000 new HCV infections in the United States in 2010, more than half (53%) involved IDU. In 2010, hepatitis B virus (HBV) infection rates were estimated to be 20% higher among people who engaged in IDU in the United States. 123


Scope of Guideline


This Practice Guideline was developed to assist in the evaluation and treatment of opioid use disorder. Although there are existing guidelines for the treatment of opioid use disorder, none have included all of the medications used for its treatment at present. Moreover, few of the existing guidelines address the needs of special populations such as pregnant women, individuals with co-occurring psychiatric disorders, individuals with pain, adolescents, or individuals involved in the criminal justice system.


Overall, the Practice Guideline contains recommendations for the evaluation and treatment of opioid use disorder, opioid withdrawal management, psychosocial treatment, special populations, and opioid overdose.



  • (1)

    Part 1: Contains guidelines on the evaluation of opioid use disorder


  • (2)

    Part 2: Provides recommendations regarding treatment options


  • (3)

    Part 3: Describes the treatment of opioid withdrawal


  • (4)

    Parts 4–6: Provide guidelines on medications for treating opioid use disorder


  • (5)

    Part 7: Describes psychosocial treatment used in conjunction with medications


  • (6)

    Parts 8–12: Provide guidelines for treating special populations and circumstances


  • (7)

    Part 13: Describes the use of naloxone in treating opioid overdose



Included and Excluded Medications


The medications covered in this guideline include the following:



  • (1)

    Methadone (Part 4)


  • (2)

    Buprenorphine (Part 5)


  • (3)

    Naltrexone in oral and extended-release injectable formulations (Part 6)


  • (4)

    Naloxone (Part 13)



All of these medications act directly upon the opioid receptors, particularly the mu-subtype. Methadone is a mu-receptor agonist; buprenorphine is a partial mu-receptor agonist; and naltrexone is an antagonist. Naloxone is a fast-acting antagonist used to reverse opioid overdose, a condition that may be life-threatening. Because of the differing actions of these medications at the receptor level, they can have very different clinical effects during treatment.


Other medications show promise for the treatment of opioid use disorder; however, there is insufficient evidence at this writing to make a full analysis of their effectiveness. For example, whereas not FDA-approved for opioid withdrawal syndrome in the United States, it is recognized that clonidine, an alpha-2 adrenergic agonist, has been in use in clinical settings for 25 years. Lofexidine (known as BritLofex, Britannia Pharmaceuticals) is approved for treating opioid withdrawal use in the United Kingdom. Because of their long history of off-label use in the United States, clonidine and buprenorphine are described for opioid withdrawal syndrome in this Practice Guideline. Again, there are other off-label medications for withdrawal management in the treatment of opioid use disorder (e.g., tramadol) that have been excluded from this guideline because there is insufficient evidence to make a full analysis of their effectiveness or consensus recommendations for their use at this time.


The ASAM recognizes that withdrawal management and withdrawal management medications could be potential topics for future guideline development. ASAM will regularly review its published guidelines to determine when partial or full updates are needed. The emergence of newly approved medications and new research will be considered as part of this process. It is also recognized that ASAM may develop guidelines or consensus documents on topics addressed in this Practice Guideline (e.g., urine drug testing). If that occurs before any update to this Practice Guideline, it is to be assumed that the recommendations in the latter documents will take precedence until this Practice Guideline is updated.


Intended Audience


This Practice Guideline is intended for all clinicians, at any level, involved in evaluating for, and/or providing, opioid use disorder treatment in the United States. The intended audience falls into the following broad groups:



  • (1)

    Physicians involved in the assessment, diagnosis, and treatment of opioid use disorder. General practice physicians (including family practitioners, pediatricians, obstetricians, and gynecologists) are often first-line providers of medical care related to opioid use disorder and are a key audience for the guideline.


  • (2)

    Clinicians involved with the completion of health assessments and delivery of health services to special populations.


  • (3)

    Clinicians involved in making an initial assessment and offering psychosocial treatments in conjunction with medications to treat opioid use disorder.


  • (4)

    Clinical case managers responsible for clinical care support, coordination of health-related and social services, and tracking of patient adherence to the treatment plan.



Qualifying Statement


The ASAM Practice Guideline is intended to aid clinicians in their clinical decision-making and patient management. It strives to identify and define clinical decision-making junctures that meet the needs of most patients in most circumstances. The ultimate judgment about care of a particular patient must be made together by the clinician and the patient in light of all the circumstances presented by the patient. As a result, situations may arise in which deviations from the Practice Guideline may be appropriate. Clinical decision-making should involve consideration of the quality and availability of expertise and services in the community wherein care is provided.


In circumstances in which the Practice Guideline is being used as the basis for regulatory or payer decisions, improvement in quality of care should be the goal. Finally, prescribed courses of treatment contained in recommendations in this Practice Guideline are effective only if the recommendations, as outlined, are followed. Because lack of patient understanding and adherence may adversely affect outcomes, clinicians should make every effort to engage the patient’s understanding of, and adherence to, prescribed and recommended pharmacological and psychosocial treatments. Patients should be informed of the risks, benefits, and alternatives to a particular treatment and should be shared parties to decision-making whenever feasible. Recommendations in this Practice Guideline do not supersede any federal or state regulation.




Methodology


Overview of Approach


These guidelines were developed using the RAND/UCLA Appropriateness Method (RAM)—a process that combines scientific evidence and clinical knowledge to determine the appropriateness of a set of clinical procedures. 64 This process is particularly appropriate for these guidelines for two reasons. First, there are very few randomized clinical trials directly comparing the approved medications for the treatment of opioid use disorder. Second, evidence supporting the efficacy of the individual medications reflects varying years of research and varying levels of evidence (e.g., nonrandomized studies, retrospective studies). The randomized clinical trial (RCT) is the gold standard for evidence-based medicine. When data are lacking from RCT, other methods must be used to help clinicians make the best choices. In addition, these guidelines are unique in that they include all three of the medications approved at present by the FDA in multiple formulations, and they address the needs of special populations such as pregnant women, individuals with pain, adolescents, individuals with co-occurring psychiatric disorder, and individuals in criminal justice. Such special populations are often excluded from RCTs, making the use of RCT data even more difficult. The RAM process combines the best available scientific evidence combined with the collective judgment of experts to yield statements about the appropriateness of specific procedures that clinicians can apply to their everyday practice.


The ASAM’s Quality Improvement Council (QIC) was the oversight committee for the guideline development. The QIC appointed a Guideline Committee to participate throughout the development process, rate treatment scenarios, and assist in writing. In selecting the committee members, the QIC made every effort to avoid actual, potential, or perceived conflicts of interest that may arise as a result of relationships with industry and other entities among members of the Guideline Committee. All QIC members, committee members, and external reviewers of the guideline were required to disclose all current related relationships, which are presented in Appendixes III, IV, and V.


The Guideline Committee comprised 10 experts and researchers from multiple disciplines, medical specialties, and subspecialties, including academic research, internal medicine, family medicine, addiction medicine, addiction psychiatry, general psychiatry, obstetrics/gynecology, and clinical neurobiology. Physicians with both allopathic and osteopathic training were represented in the Guideline Committee. The Guideline Committee was assisted by a technical team of researchers from the Treatment Research Institute (TRI) affiliated with the University of Pennsylvania, and worked under the guidance of Dr. Kyle Kampman who led the TRI team as Principal Investigator in implementing the RAM.


The RAM process is a deliberate approach encompassing review of existing guidelines, literature reviews, appropriateness ratings, necessity reviews, and document development. The steps are summarized in the flow chart in “Exhibit 1: Methodology.”


Task 1: Review of Existing Guidelines


Review of Existing Clinical Guidelines


All existing clinical guidelines that addressed the use of medications and psychosocial treatments in the treatment of opioid use disorders including special populations (e.g., pregnant women, individuals with pain, and adolescents), and that were published during the period from January 2000 to April 2014, were identified and reviewed. In total, 49 guidelines were identified and 34 were ultimately included in the analysis. See “Appendix I” for a list of the guidelines that were reviewed. The included guidelines offered evidence-based recommendations for the treatment of opioid use disorder using methadone, buprenorphine, naltrexone, and/or naloxone.


The majority of existing clinical guidelines are based on systematic reviews of the literature including appropriateness criteria used in the RAM. Therefore, the aim of this exercise was not to re-review all of the research literature, but to identify within the existing clinical guidelines how they addressed common questions or considerations that clinicians are likely to raise in the course of deciding whether and how to use medications as part of the treatment of individuals with opioid use disorder.


Analysis of Clinical Guidelines


On the basis of the previously reviewed existing clinical guidelines, an analytic table was created and populated to display the identified key components. This table served as the foundation for development of hypothetical statements. The hypothetical statements were sentences describing recommendations derived from the analysis of the clinical guidelines.


Preparation of Literature Review on Psychosocial Interventions


A review of the literature on the efficacy of psychosocial treatment delivered in conjunction with medications for the treatment of opioid use disorder was conducted. This review was partially supported by funding from the National Institute on Drug Abuse (NIDA). Articles were identified for inclusion in the review through searches conducted in two bibliographic databases (e.g., PsycINFO and PubMed) using predefined search terms and established selection criteria. Titles and abstracts were reviewed for inclusion by two members of the research team.


To increase the overall relevance of the review, the search was limited to articles in the 6-year period from 2008 to the present. In the event that the article reflected a secondary analysis of data from a relevant study, the original study was included in the literature review. In addition, findings from three prominent systematic reviews (i.e., 2007 review on psychosocial interventions in pharmacotherapy of opioid dependence prepared for the Technical Development Group for the World Health Organization, “Guidelines for Psychosocially Assisted Pharmacotherapy of Opioid Dependence,” and two 2011 Cochrane reviews examining psychosocial and pharmacological treatments for opioid withdrawal management and psychosocial interventions combined with agonist treatment) were summarized. 4,5,55


The literature search yielded 938 articles. The titles and abstracts were reviewed to determine if the study met the inclusion/exclusion criteria, and those that did not ( n = 787) were removed. The remaining 151 articles were then reviewed for inclusion, and 27 articles were ultimately retained for use in the literature review, as the others did not meet the predetermined inclusion/exclusion criteria. These articles, along with the relevant systematic reviews of the literature, are described in the literature review in the next section.


Task 2: Identification of Hypothetical Statements and Appropriateness Rating


RAND/UCLA Appropriateness Method


The first step in the RAM is to develop a set of hypothetical statements, which were derived from the guideline analysis and literature review described in the previous section, for appropriateness rating.


The analysis and literature review generated a list of 245 hypothetical statements that reflected recommended medical or psychosocial treatment. Each member of the Guideline Committee reviewed the guideline analysis and literature review, and privately rated 245 hypothetical clinical statements on a nine-point scale of “appropriateness.” In the context of this Practice Guideline, the meaning of appropriateness was defined as:


A statement, procedure or treatment is considered to be appropriate if the expected health benefit (e.g., increased life expectancy, relief of pain, reduction in anxiety, improved functional capacity) exceeds the expected negative consequences (e.g., mortality, morbidity, anxiety, pain) by a sufficiently wide margin that the procedure is worth doing, exclusive of cost.


An appropriateness score of 1 meant that the statement was “highly inappropriate.” An appropriateness rating of 9 meant that the statement was “highly appropriate.” These appropriateness statements were meant to identify a lack of consensus in existing guidelines and research literature.


Guideline Committee Meeting


Upon completion and collection of the individual Guideline Committee member ratings, 201 of the 245 hypothetical statements were identified as meeting the criteria for consensus. The remaining 44 statements had divergent ratings. On September 15, 2014, the Guideline Committee met in Washington, District of Columbia, to discuss the hypothetical clinical statements. At this meeting, the committee came to consensus on the hypothetical statements. After the meeting, the information gathered was used to revise several of the statements; and the Guideline Committee was asked to re-rate the revised statements.


Literature Review


A supplementary literature review was also conducted to identify relevant studies that might resolve statements that had resulted in divergent ratings during the Guideline Committee meeting. Information relating to the vast majority of these divergent ratings was subsequently found within the existing guideline data set, and consequently included in the first draft of the Practice Guideline.


For the topics and questions for which answers were not found in the existing guideline data set, a full literature review was conducted. The topics and questions for which no further clarification was found in the literature were considered “gaps” that require additional research before inclusion in this guideline. These gaps in the literature were: urine drug testing; patients using marijuana; the safety of delivering injectable naltrexone doses to patients with high metabolism every 3 weeks; and the safety of adding full agonists to treatment with buprenorphine for pain management.


Creation and Revision of Guideline Outline


All the identified appropriate/uncertain hypothetical statements and supporting research were incorporated into an outline defining each specific section to be included in the final Practice Guideline. The draft outline, review of existing guidelines, and literature review were all sent to the Guideline Committee members for review and discussion during two web teleconferences and through private communication. Two teleconferences were held to ensure full participation from members of the Guideline Committee.


Task 3: Comparative Analysis, Review, and Necessity Rating


Committee Review and Rating


The Guideline Committee then re-rated the 211 “appropriate” hypothetical statements for necessity. When rating for necessity, the Guideline Committee members were asked to adhere to the following guidance:


A statement was considered necessary when all the following criteria were met:



  • (1)

    It would be considered improper care not to provide this service.


  • (2)

    Reasonable chance exists that this procedure and/or service will benefit the patient. (A procedure could be appropriate if it had a low likelihood of benefit, but few risks; however, such procedures would not be necessary.)


  • (3)

    The benefit to the patient is of significance and certainty. (A procedure could be appropriate if it had a minor but almost certain benefit, but it would not be necessary.)



Necessity is a more stringent criterion than appropriateness. If a procedure is necessary, this means that the expected benefits outweigh the expected harms (i.e., it is appropriate), and that they do so by such a margin that the physician must recommend the service. Of course, patients may decline to follow their physician’s recommendations. 64


Of the 211 rated statements, 184 hypothetical statements met the criteria for being both appropriate and necessary, and were incorporated in the guideline.


Final Draft Outline


The final draft outline highlighted hypothetical statements that had been determined to rise to the level of necessity.


Task 4: Drafting the National Practice Guideline


Draft and Review


A first draft of the Practice Guideline was created using the Guideline Committee’s recommendations resulting from supporting evidence and the appropriateness and necessity ratings discussed above. The first draft of the Practice Guideline was sent to the Guideline Committee for review and electronic comment. During a subsequent teleconference in January 2015, the Guideline Committee discussed the comments received via first review. Revisions were made to the draft, which went again through subsequent reviews by the Guideline Committee and the ASAM Quality Council throughout February and March 2015.


Task 5: External Review


External Review


The ASAM sought input from ASAM members—patients and caregiver groups, stakeholders including experts from criminal justice system, government agencies, other professional societies, and hospitals and health systems. ASAM also made the document and a qualitative review guide available to ASAM members and the general public for a one week period of review and comment. The final draft Practice Guideline was submitted to the ASAM Board of Directors in April 2015.




Exhibit 1: Methodology






Part 1: Assessment and Diagnosis of Opioid Use Disorder


Comprehensive Assessment


The ASAM Standards of Care for the Addiction Specialist Physician (the “ASAM Standards”) describe the importance of comprehensive assessment. Although the assessment process is ongoing for the patient with substance use disorder, a comprehensive assessment is “a critical aspect of patient engagement and treatment planning” and should be conducted during the initial phase of treatment. 118


The assessment is not necessarily the first visit; it is critical, however, to determine emergent or urgent medical problems. Patients with opioid use disorder often have other physiological or psychiatric conditions that may complicate their treatment. These concomitant medical and psychiatric conditions may need immediate attention and require transfer to a higher level of care (see “Part 11: Special Populations: Individuals With Co-occurring Psychiatric Disorders.”)


Medical History


The patient’s medical history should include screening for concomitant medical conditions and routine identification of medications, allergies, pregnancy, family medical history, and so on. Particular attention should be paid to the following: history of infectious diseases such as hepatitis, HIV, and TB; acute trauma; psychiatric, substance use, addictive behavior, and addiction treatment history; and any previous history of pharmacotherapy. An intake of the patient’s social history and assessment of readiness for change including identification of any facilitators and barriers are also components of the medical history.


Physical Examination


As part of the comprehensive assessment of patients with opioid use disorder, a physical examination should be completed by the prescriber him/herself (the clinician authorizing the use of a medication for the treatment of opioid use disorder), another member of the clinician’s health system, or the prescribing physician. Furthermore, the responsible clinician should assure that a current physical examination (in accordance with the ASAM Standards) is contained within the patient medical record before a patient is started on a new medication for the treatment of his/her opioid use disorder.


The examination should include identifying objective physical signs of opioid intoxication or withdrawal. See Table 1 for a list of common signs of intoxication or withdrawal. In addition, the examination should evaluate objective signs of substance use disorders. See Table 2 for a list of physical signs of substance use disorders (including opioid use disorder).



Table A3.1

Common Signs of Opioid Intoxication and Withdrawal.










Intoxication Signs Withdrawal Signs
Drooping eyelids
Constricted pupils
Reduced respiratory rate
Scratching (due to histamine release)
Head nodding
Restlessness, irritability, anxiety
Insomnia
Yawning
Abdominal cramps, diarrhea, vomiting
Dilated pupils
Sweating
Piloerection


Table A3.2

Objective Physical Signs in Substance Use Disorders.










System Findings
Dermatologic
Ear, nose, throat, and eyes
Mouth
Cardiovascular
Respiratory
Musculoskeletal and extremities
Gastrointestinal
Abscesses, rashes, cellulitis, thrombosed veins, jaundice, scars, track marks, pock marks from skin popping
Pupils pinpoint or dilated, yellow sclera, conjunctivitis, ruptured eardrums, otitis media, discharge from ears, rhinorrhea, rhinitis, excoriation or perforation of nasal septum, epistaxis, sinusitis, hoarseness, or laryngitis
Poor dentition, gum disease, abscesses
Murmurs, arrhythmias
Asthma, dyspnea, rales, chronic cough, hematemesis
Pitting edema, broken bones, traumatic amputations, burns on fingers
Hepatomegaly, hernias


Table A3.3

Contraindications and Precautions for Pharmacotherapy Options.
































Medication Contraindications Warnings and Precautions
Methadone Hypersensitivity
Respiratory depression
Severe bronchial asthma or hypercapnia
Paralytic ileus
Cardiac conduction effects
Diversion and misuse are possible
Physical dependence
Respiratory depression when used in association with CNS depressants including alcohol, other opioids, and illicit drugs
Head injury and increased intracranial pressure
Liver disease
Respiratory insufficiency
Concomitant substance use disorders
Co-occurring psychiatric disorders
Drug interactions with medications metabolized by cytochrome P450 enzymes principally CYP34A, CYP2B6, CYP2C19, and to a lesser extent by CYP2C9 and CYP2D6 drugs coadministered with methadone should be evaluated for interaction potential
Buprenorphine(all formulations) Hypersensitivity Diversion and misuse are possible
Physical dependence
Respiratory depression when used in association with CNS depressants including alcohol, other opioids, and illicit drugs
Precipitated withdrawal if used in patients physically dependent on full agonists opioids before the agonist effects have worn offNeonatal withdrawal has been reported after use of buprenorphine during pregnancy
Not recommended for patients with severe hepatic impairment
May cause sedation
Naltrexone (oral and injectable formulations) Hypersensitivity reactions to naltrexone, or for injectable previous hypersensitivity reactions to polylactide-co-glycolide, carboxymethylcellulose, or any other constituent of the diluent Vulnerability to overdose
Injection site reactions associated with injectable naltrexone
Precipitated opioid withdrawal
Risk of hepatotoxicity
Patients currently physically dependent on opioids, including partial agonists Patient should be monitored for the development of depression and suicidality
Patients receiving opioid analgesics
Patients in acute opioid withdrawal
Emergency reversal of opiate blockade may require special monitoring in a critical care setting
Eosinophil pneumonia has been reported in association with injectable naltrexone
Administer IM injections with caution to patients with thrombocytopenia or a coagulation disorder

IM, Intramuscular.


Special attention should be given to identifying IDU by the presence of new or older puncture marks. Common injection sites are inside the elbow (cubital fossa) and forearm, but other sites on the extremities may be injection sites.


Assessment and History Considerations Specific to Females


Use of contraception and determination of pregnancy are factors in choosing treatment options for women with opioid use disorder. Contraception and reproductive health are topics of discussion within the assessment process of female patients who are considering opioid use disorder treatment. Clinicians and female patients should keep in mind that fertility increases as treatment becomes effective. Case management plans may need to include referral to gynecological services for female patients. An in-depth discussion of the treatment of opioid use disorder in pregnant women is described later in “Part 8: Special Populations: Pregnant Women.”


Laboratory Tests


Initial lab testing should include hepatitis C and HIV testing. Hepatitis serology and vaccination are recommended. Hepatitis A and B testing and vaccination should be offered when appropriate. As above, women of childbearing potential and age should be tested for pregnancy. Tuberculosis testing and testing for sexually transmitted infections, including syphilis, may be considered.


A complete blood count and liver function study should be conducted to screen for liver dysfunction, infection, and other medical conditions. Abnormal results may require further investigation.


Assessment for Mental Health Status and Psychiatric Disorder


Patients being evaluated for opioid use disorder, and/or for possible medication use in the treatment of opioid use disorder, should undergo an evaluation of possible co-occurring psychiatric disorders. During the assessment process and physical examination, it is important for the clinician to assess for mental health status consistent with the ASAM Standards.


In the ASAM Standards, I.1 indicates that the physician “assures that an initial comprehensive, multicomponent assessment is performed for each patient, either by performing it her/himself or by assuring it is conducted in full or in part by another qualified professional within the system in which she/he is working.” A thorough medical and psychiatric history and family history is indicated as a component of this same standard. Patients who are determined as exhibiting urgent or emergent psychiatric conditions, or who are psychiatrically unstable and represent a danger to themselves or others, should be referred to the appropriate level of care for their safety and the safety of others. Further specialty evaluation may be warranted depending on severity of indicators for psychiatric instability. Indicators of psychiatric instability or disorder include acute suicidal or homicidal ideation, acute psychosis, and delirium.


Assessment for Alcohol and Substance Use and Treatment History


A careful evaluation of current and past use of alcohol and drugs, including nonmedical use of prescription medications, is required to diagnose opioid use disorder. Because opioid use disorder may co-occur with other use disorders, the evaluator should assess frequency and quantity of use.


Completing a history of opioid drug use with a patient who has been identified as using opioids should focus on the following:



  • (1)

    type and amount of opioid(s) used recently;


  • (2)

    route of administration;


  • (3)

    last use;


  • (4)

    treatment history; and


  • (5)

    problems resulting from drug use.



The amount of drug being consumed will impact the likelihood and severity of withdrawal symptoms when the drug is stopped, so it is useful to obtain an estimate of the amount used (each time and number of times per day).


Prescription Drug Monitoring Programs (PDMPs) offer information about prescription opioid use. They can serve as important resources for clinicians’ use in completing full patient clinical assessments of opiate and other controlled substance use history, and it is recommended that they be utilized. It is recognized, as detailed in “Exhibit 2: Prescription Drug Monitoring Programs,” that there is variation across states in terms of the level of operation of these programs, the extent of their data sharing across states, and state requirements for their use before prescribing controlled substances.


In addition, a history of outpatient and inpatient treatment for alcohol and other substance use disorders should be collected. Clinicians should ask for information about the type and duration of treatment and outcomes.


Assessment for Co-Occurring Alcohol and Substance Use


Opioid use disorder often co-occurs with alcohol and other substance use disorders. Therefore, evaluation of co-occurring alcohol and substance use is recommended.


Clinicians should assess signs and symptoms of alcohol or sedative, hypnotic, or anxiolytic intoxication or withdrawal. Alcohol or sedative, hypnotic, or anxiolytic withdrawal may result in seizures, hallucinosis, or delirium, and may represent a medical emergency. Likewise, concomitant use of alcohol and sedatives, hypnotics, or anxiolytics with opioids may contribute to respiratory depression. Patients with significant co-occurring substance use disorders, especially severe alcohol or sedative, hypnotic, or anxiolytic use, may require a higher level of care.


An evaluation of past and current substance use should be conducted, and a determination as to whether addiction involving other substances or other behaviors is present. For instance, the regular use of marijuana or cannabinoids, tobacco or electronic nicotine delivery devices, or other drugs should not be a reason to suspend medication use in the treatment of addiction involving opioid use. Concurrent use of other drugs or active engagement in other addictive behaviors should lead to consideration of other treatment plan components for the patient. The presence of co-occurring substance use disorders should provoke a re-evaluation of the level of care that is in place for psychosocial treatment, along with pharmacological therapy. In most cases, co-occurring drug use will not represent a medical emergency. In such cases, patients can be treated for both their opioid use disorder and co-occurring alcohol or substance use disorders. However, ongoing use of other drugs may lead to poorer treatment outcomes. Evidence does demonstrate that individuals who are actively using other substances during opioid use disorder treatment have a poorer prognosis. 66,99,128


The Guideline Committee cautioned against excluding patients from treatment for their opioid use disorder because they are using marijuana or other psychoactive substances that do not interact with opioids, and that are not prescribed by their physician. Whereas there is a paucity of research examining this topic, evidence demonstrates that patients under treatment have better outcomes than those not retained under treatment. 82,108


Suspension of opioid use disorder treatment may increase the risk for death from overdose, accidents, or other health problems. However, continued use of marijuana or other psychoactive substances may impede treatment for opioid use disorder; thus, an approach emphasizing cessation of all unprescribed substances is likely to result in the best results. Further research is needed on the outcomes of patients in opioid use disorder treatment who are continuing the nonmedical use of psychoactive substances.


Assessment for Tobacco Use


Tobacco use should be queried, and the benefits of cessation should be promoted routinely with patients presenting for evaluation and treatment of opioid use disorder. Several studies have demonstrated that smoking cessation improves long-term outcomes among individuals receiving treatment for substance use disorders. 15,129,157


Assessment of Social and Environmental Factors


Clinicians should conduct an assessment of social and environmental factors (as outlined in the ASAM Standards) to identify facilitators and barriers to addiction treatment and specifically to pharmacotherapy. Before a decision is made to initiate a course of pharmacotherapy for the patient with opioid use disorder, the patient should receive a multidimensional assessment in fidelity with the ASAM Criteria: Treatment Criteria for Addictive, Substance-Related, and Co-occurring Conditions (the “ASAM Criteria”). The ASAM Patient Placement Criteria uses six dimensions to create a holistic biopsychosocial assessment of an individual to be used for service planning and treatment. Dimension one is acute intoxication or withdrawal potential. Dimension two is biomedical conditions and conditions. Dimension three is emotional, behavioral, or cognitive conditions or complications. Dimension four is readiness for change. Dimension five is continued use or continued problem potential. Dimension six is recovery/living environment. 111


The use of medications for the patient with addiction involving opioid use can be appropriate across all levels of care. Pharmacotherapy is not a “level of care” in addiction treatment, but one component of multidisciplinary treatment. Whereas medication as a standalone intervention has been utilized in North America and internationally, ASAM recommends that the use of medications in the treatment of addiction be part of a broad bio-psycho-social-spiritual intervention appropriate to the patient’s needs and to the resources available in the patient’s community. Addiction should be considered a bio-psycho-social-spiritual illness, for which the use of medication(s) is but only one component of overall treatment.




Exhibit 2: Prescription Drug Monitoring Programs




Diagnosing Opioid Use Disorder


Opioid use disorder is primarily diagnosed on the basis of the history provided by the patient and a comprehensive assessment that includes a physical examination. Corroborating information reported by significant others can be used to confirm the diagnosis, especially when there is lack of clarity or inconsistency in information. Other clinicians may make a diagnosis of opioid use disorder; however, provider confirmation of the diagnosis is required before medications are prescribed. This is discussed further in later parts that address specific medications.


DSM-5 Criteria for Diagnosis


The diagnosis of opioid use disorder is based on criteria outlined in the DSM-5. The criteria describe a problematic pattern of opioid use leading to clinically significant impairment or distress. There are a total of 11 symptoms and severity is specified as either mild (presence of 2–3 symptoms), moderate (presence of 4–5 symptoms) or severe (presence of 6 or more symptoms) within a 12-month period. Opioid use disorder requires that at least two of the following 11 criteria be met within a 12-month period: (1) taking opioids in larger amounts or over a longer period of time than intended; (2) having a persistent desire or unsuccessful attempts to reduce or control opioid use; (3) spending excess time obtaining, using, or recovering from opioids; (4) craving for opioids; (5) continuing opioid use causing inability to fulfill work, home, or school responsibilities; (6) continuing opioid use despite having persistent social or interpersonal problems; (7) lack of involvement in social, occupational, or recreational activities; (8) using opioids in physically hazardous situations; (9) continuing opioid use in spite of awareness of persistent physical or psychological problems; (10) tolerance, including need for increased amounts of opioids or diminished effect with continued use at the same amount—as long as the patient is not taking opioids under medical supervision; and (11) withdrawal manifested by characteristic opioid withdrawal syndrome or taking opioids to relieve or avoid withdrawal symptoms—as long as the patient is not taking opioids under medical supervision.


More detail about diagnosing opioid use disorder is available in the American Psychiatric Association: Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition . Arlington, VA, American Psychiatric Association, 2013.


Withdrawal Scales


There are a number of useful opioid withdrawal scales that can assist the clinician in evaluating patients with opioid use disorder by identifying and quantitating the severity of opioid withdrawal symptoms. The Objective Opioid Withdrawal Scale (OOWS), which relies on clinical observation, is useful in measuring and documenting the objectively measurable symptoms of opioid withdrawal. The Subjective Opioid Withdrawal Scale (SOWS) records the patient’s rating of opioid withdrawal on a 16-item scale. 74


Finally, the Clinical Opioid Withdrawal Scale (COWS) includes 11 items, and contains signs and symptoms of opioid withdrawal, which are both objective and subjective in nature. 74


Urine Drug Testing


Urine drug testing, or other reliable biological tests for the presence of drugs, during the initial evaluation and frequently throughout treatment, is highly recommended. There are a variety of toxicology tests available, some with greater and lesser reliability and validity. The person who is interpreting these labs should be very familiar with the methodology and the reliability. There is little research on the optimal frequency of testing. The recommendations given below are based on the consensus opinion of the Guideline Committee. The frequency of drug testing will be determined by a number of factors, including the stability of the patient, the type of treatment, the treatment setting, and the half-life of drugs in the matrix being tested. Patients will likely require more testing early in treatment or during periods of relapse. Patients participating in office-based treatment with buprenorphine may be tested at each office visit. Patients participating in treatment for opioid use disorder at Opioid Treatment Programs (OTPs) are mandated by the Federal law 30 to receive a minimum of eight drug tests per year, but may be tested more frequently based on clinical need. More detailed information on drug testing is contained in “Drug Testing: A White Paper of the American Society of Addiction Medicine.” 10


Opioids are detectable in the urine for 1–3 days after use. A negative urine test combined with no history of withdrawal may indicate a lack of physical dependence. However, a negative urine test does not rule out opioid use, disorder, or physical dependence. Urine testing is also helpful to identify use of other psychoactive substances.


Summary of Recommendations


Assessment Recommendations




  • (1)

    First clinical priority should be given to identifying and making appropriate referral for any urgent or emergent medical or psychiatric problem(s), including drug-related impairment or overdose.


  • (2)

    Completion of the patient’s medical history should include screening for concomitant medical conditions including infectious diseases (hepatitis, HIV, and TB), acute trauma, and pregnancy.


  • (3)

    A physical examination should be completed as a component of the comprehensive assessment process. The prescriber (the clinician authorizing the use of a medication for the treatment of opioid use disorder) may conduct this physical examination him/herself, or, in accordance with the ASAM Standards, ensure that a current physical examination is contained within the patient medical record before a patient is started on a new medication for the treatment of his/her addiction.


  • (4)

    Initial laboratory testing should include a complete blood count, liver function tests, and tests for hepatitis C and HIV. Testing for TB and sexually transmitted infections should also be considered. Hepatitis B vaccination should be offered, if appropriate.


  • (5)

    The assessment of women presents special considerations regarding their reproductive health. Women of childbearing age should be tested for pregnancy, and all women of childbearing potential and age should be queried regarding methods of contraception, given the increase in fertility that results from effective opioid use disorder treatment.


  • (6)

    Patients being evaluated for addiction involving opioid use, and/or for possible medication use in the treatment of opioid use disorder, should undergo (or have completed) an assessment of mental health status and possible psychiatric disorders (as outlined in the ASAM Standards).


  • (7)

    Opioid use is often co-occurring with other substance-related disorders. An evaluation of past and current substance use and a determination of the totality of substances that surround the addiction should be conducted.


  • (8)

    The use of marijuana, stimulants, or other addictive drugs should not be a reason to suspend opioid use disorder treatment. However, evidence demonstrates that patients who are actively using substances during opioid use disorder treatment have a poorer prognosis. The use of benzodiazepines and other sedative hypnotics may be a reason to suspend agonist treatment because of safety concerns related to respiratory depression.


  • (9)

    A tobacco use query and counseling on cessation of tobacco products and electronic nicotine delivery devices should be completed routinely for all patients, including those who present for evaluation and treatment of opioid use disorder.


  • (10)

    An assessment of social and environmental factors should be conducted (as outlined in the ASAM Standards to identify facilitators and barriers to addiction treatment, and specifically to pharmacotherapy). Before a decision is made to initiate a course of pharmacotherapy for the patient with opioid use disorder, the patient should receive a multidimensional assessment in fidelity with the ASAM Criteria. Addiction should be considered a bio-psycho-social-spiritual illness, for which the use of medication(s) is but only one component of overall treatment.



Diagnosis Recommendations




  • (1)

    Other clinicians may diagnose opioid use disorder, but confirmation of the diagnosis by the provider with prescribing authority and who recommends medication use must be obtained before pharmacotherapy for opioid use disorder commences.


  • (2)

    Opioid use disorder is primarily diagnosed on the basis of the history provided by the patient and a comprehensive assessment that includes a physical examination.


  • (3)

    Validated clinical scales that measure withdrawal symptoms, for example, the OOWS, SOWS, and the COWS, may be used to assist in the evaluation of patients with opioid use disorder.


  • (4)

    Urine drug testing during the comprehensive assessment process, and frequently during treatment, is recommended. The frequency of drug testing is determined by a number of factors, including the stability of the patient, the type of treatment, and the treatment setting.



Areas for Further Research




  • (1)

    More research is needed on best practices for drug testing during the initial evaluation and throughout the entire treatment process.


  • (2)

    Further research is needed on evidence-based approaches for treating opioid use disorder in patients who continue to use marijuana and/or other psychoactive substances.


  • (3)

    Whereas research indicates that offering tobacco cessation is a standard for all medical care, more research is needed before specific evidence-based recommendations can be made.





Part 2: Treatment Options


Introduction


Once the diagnosis of opioid use disorder has been established, and it has been determined that the patient is medically and psychiatrically stable, the next task is to decide on a course of treatment. Potential treatments include withdrawal management in conjunction with psychosocial treatment, or psychosocial treatment combined with one of three medications: methadone, buprenorphine, or naltrexone (oral or extended-release injectable formulations). Withdrawal management alone can be the first step, but is not a primary treatment for opioid use disorder and should “only” be considered as a part of a comprehensive and longitudinal plan of care that includes psychosocial treatment, with or without medication-assisted therapy.


The choice among available treatment options should be a shared decision between the clinician and the patient. There are a number of factors to consider in deciding what treatment to choose. Among the first considerations are the priorities of the patient, for instance: Is the patient open to pharmacotherapy? What type of treatment setting does the patient prefer? Does the patient understand the physical dependence aspects of treatment medication? A patient’s past experiences with treatment for opioid use disorder should be considered as well. Of course, above all, evidence supporting the potential efficacy and safety of the various treatments is critically important.


For most patients with opioid use disorder, the use of medications (combined with psychosocial treatment) is superior to withdrawal management (combined with psychosocial treatment), followed finally by psychosocial treatment on its own. This is true for both agonist and partial agonist, and antagonist medications. Evidence suggests that methadone maintenance treatment is superior to withdrawal management alone and significantly reduces opioid drug use. 107


Further, mortality is lower in patients on methadone, as compared to those not undergoing treatment. 50 Methadone also lowers the risk of acquiring or spreading HIV infection. 162,124 In clinical studies, evidence favors buprenorphine, compared to no treatment, in decreasing heroin use and improving treatment retention. 82,98 Finally, evidence supports the efficacy of both oral naltrexone and extended-release injectable naltrexone versus placebo for the treatment of opioid use disorder. 40,92,154


Pharmacotherapy Options


The medications covered in this guideline are mainly those that have been approved by the FDA for the treatment of opioid dependence as defined in prior versions of the DSM-III and DSM-IV, and “not necessarily” the definition contained in the current version of the manual, the DSM-5. DSM-5 combined “opioid abuse” and “opioid dependence” criteria from prior versions of the DSM and included them in the new definition of “opioid use disorder.” As a result, pharmacologic treatment may not be appropriate for all patients along the entire opioid use disorder continuum. In a study comparing opioid dependence from DSM-IV and opioid use disorder from DSM-5, optimal concordance occurred when four or more DSM-5 criteria were endorsed (i.e., the DSM-5 threshold for moderate opioid use disorder). 42


The medications discussed in this Practice Guideline all have ample evidence supporting their safety and efficacy. It is recognized that other medications have been used off-label to treat opioid use disorder, but with some exceptions (clearly noted in the text) the Guideline Committee has not issued recommendations on the use of these medications. Cost-efficacy was not a consideration in the development of this Practice Guideline.


Each medication will be discussed in detail in subsequent sections:



  • (1)

    Methadone (mu-agonist) for opioid use disorder treatment and withdrawal management (Part 4).


  • (2)

    Buprenorphine (partial mu-agonist) for opioid use disorder treatment and withdrawal management (Part 5).


  • (3)

    Naltrexone (antagonist) for relapse prevention (Part 6).


  • (4)

    Naloxone (antagonist) to treat overdose (Part 13).



The only medication that is “not” FDA-approved for the treatment of opioid use disorder that will be covered in this Practice Guideline is the use of the alpha-2 adrenergic agonist, clonidine, for the treatment of opioid withdrawal (see “Part 3: Treating Opioid Withdrawal”).


Key outcomes in evaluating the efficacy of the various pharmacotherapies include: decreased mortality, abstinence from opioids, and retention in treatment. In regards to these key outcomes, there is some evidence supporting the relative efficacy of one medication over another, but in many cases, there are no good-quality studies comparing the relative benefits of one medication over another. As noted above, there is strong evidence supporting the superiority of methadone over drug-free treatment for reducing mortality, reducing opioid use, and promoting treatment retention. 138


Efficacy Considerations


Treatment Setting


In accordance with US Federal laws and regulations derived from the Harrison Act and Congressional exceptions to that 1914 law, the venue in which treatment for opioid use disorder is provided is as important a consideration as is the specific medication selected (methadone vs. buprenorphine vs. naltrexone). 75 Federal and state-licensed OTPs offer daily supervised dosing of methadone. OTPs are state and federally regulated to dispense opioid agonist treatment. An increasing number of such highly regulated programs also offer the option of daily supervised dosing of buprenorphine.


In accordance with Federal law 21 CFR §1306.07, office-based opioid treatment (OBOT), which provides authorization of medication via regular outpatient prescriptions filled in a retail pharmacy like any other prescription medication, is available for buprenorphine, but not for methadone. Physicians in private practices, or various other types of private and public sector clinics, can be authorized to prescribe outpatient supplies of the partial opioid agonist buprenorphine. This flexibility to provide OBOT is discussed more in “Part 5: Buprenorphine.” There are no regulations regarding facilities themselves, but rather of the individual physician who prescribes buprenorphine (see “Part 5: Buprenorphine” for physician qualifications associated with OBOT).


Naltrexone can be prescribed in any setting by any clinician with the authority to prescribe any medication. It is not listed among federal or state-controlled substances schedules, and there are no regulations of facilities or prescribers for the use of naltrexone in the treatment of opioid use disorder (such that there are for OTP and OBOT).


It is recommended that the clinician consider a patient’s psychosocial situation, co-occurring disorders, and opportunities for treatment retention versus risks of diversion when determining whether OTP or OBOT is most appropriate.


Pharmacology


Differences in efficacy may also arise from differences in pharmacology; whereas methadone is a full agonist at the mu-opioid receptor and produces higher levels of physiological dependence; buprenorphine is a partial agonist with less physiological dependence. There are few studies comparing the relative efficacy of methadone versus buprenorphine in reducing opioid use. Likewise, evidence supports the efficacy of naltrexone for relapse prevention compared to a placebo control. 40,93 There is an absence of studies that compare treatment using either oral naltrexone or extended-release injectable naltrexone versus agonist treatment with either methadone or buprenorphine.


Contraindications and Precautions


The following section describes the major indications, contraindications, and precautions for methadone, buprenorphine, and naltrexone. This section is a summary and is not an exhaustive description of medication information. (Refer to Table 3 below for a summary of contraindications and precautions.)


Methadone


Methadone is frequently used to manage withdrawal symptoms from opioids and is recommended for pharmacological treatment of opioid use disorder (see “Part 4: Methadone”).


Methadone is “contraindicated” for the following conditions:



  • (1)

    Patients with known hypersensitivity to methadone hydrochloride.


  • (2)

    Patients experiencing respiratory depression (in the absence of resuscitative equipment or in unmonitored settings).


  • (3)

    Patients with acute bronchial asthma or hypercapnia (also known as hypercarbia).


  • (4)

    Patients with known or suspected paralytic ileus.



Methadone should be used with “caution” for the following conditions:



  • (1)

    Patients with decompensated liver disease (e.g., jaundice, ascites) due to increased risk of hepatic encephalopathy.


  • (2)

    Patients with respiratory insufficiency.


  • (3)

    Patients with concomitant substance use disorders, particularly patients with sedative, hypnotic, or anxiolytic use disorders. Interactions between methadone and hypnotics, sedatives, or anxiolytics may be life-threatening.


  • (4)

    Patients with concomitant psychiatric diagnoses that impair their ability to maintain daily attendance at an OTP.


  • (5)

    Patients with low levels of physical dependence to opioids should be started with low doses of methadone.



Significant “medication interactions” to consider before starting methadone are as follows:



  • (1)

    Methadone may prolong the QT interval and should be used in caution with other agents that may also prolong the QT interval. These include class I or class III anti-arrhythmic drugs, calcium channel blockers, some anti-psychotics, and some antidepressants.


  • (2)

    Methadone is metabolized through the cytochrome P450 enzyme pathway. Many agents interact with this pathway including alcohol, anticonvulsants, antiretrovirals, and macrolide antibiotics.



Buprenorphine


Buprenorphine is a partial opioid agonist and mixed opioid agonist–antagonist. It is usually provided in a formulation that includes naloxone. Buprenorphine is recommended for pharmacological treatment of opioid use disorder (see “Part 5: Buprenorphine”).


Buprenorphine is also an effective treatment for opioid withdrawal with efficacy similar to methadone, and much superior to clonidine in opioid withdrawal management. 20,35,97


Although one trial did find that longer courses of buprenorphine with gradual tapering were superior to rapid tapering for withdrawal, 137 there is insufficient evidence on outcomes to make recommendations on buprenorphine taper duration.


Buprenorphine is “contraindicated” for the following conditions:



  • (1)

    Patients with hypersensitivity to buprenorphine or any component of the formulation.


  • (2)

    Patients with severe liver impairment are not good candidates for office-based treatment with buprenorphine. (Patients with hepatitis C infection who do not have severe liver impairment may, however, be considered for buprenorphine.)



Buprenorphine should be used with “caution” for the following conditions:



  • (1)

    Patients in whom hepatitis has been reported, particularly in patients with previous hepatic dysfunction. A direct comparison of the effects of buprenorphine and methadone, however, showed no evidence of liver damage during the initial 6 months in either treatment groups. 134 Monitoring liver function in patients at increased risk for hepatotoxicity may be considered.


  • (2)

    Patients who, at present, have an alcohol use or sedative, hypnotic, or anxiolytic use disorder.


  • (3)

    Patients with hypovolemia, severe cardiovascular disease, or taking drugs that may exaggerate hypotensive effects. Buprenorphine may cause hypotension, including orthostatic hypotension and syncope.



Significant “medication interactions” to consider before starting buprenorphine include the following:



  • (1)

    Alcohol and sedatives, hypnotics, or anxiolytics may enhance the central nervous system depressive effect of buprenorphine.


  • (2)

    Buprenorphine is metabolized to norbuprenorphine primarily by cytochrome CYP3A4; therefore, potential interactions may occur when buprenorphine is given concurrently with agents that affect CYP3A4 activity. The concomitant use of buprenorphine with CYP3A4 inhibitors (e.g., azole antifungals such as ketoconazole, macrolide antibiotics such as erythromycin, and HIV protease inhibitors) should be monitored and may require dose reduction of one or both agents. 27,144,173



Naltrexone


Naltrexone is recommended for pharmacological treatment of opioid use disorder (see “Part 6: Naltrexone”). Naltrexone is an opioid antagonist that blocks the effects of opioids. It is a pharmacotherapy option used to treat opioid use disorder and prevent relapse after detoxification. Naltrexone causes immediate withdrawal symptoms (precipitated withdrawal) in a person with active physical dependence on opioids. There are oral and extended-release injectable formulas of naltrexone. Oral naltrexone, if taken daily, is most effective in patients who are highly motivated or legally mandated to receive treatment, and/or when taking the medication is closely supervised. Conversely, the efficacy of oral naltrexone for the treatment of opioid use disorder is often adversely affected by poor medication adherence. 114 Clinicians may want to reserve using oral naltrexone for patients who are able to comply with special techniques to enhance their adherence, for example, observed dosing. An extended-release injectable naltrexone formulation is available, which may overcome the adherence limitations of the oral formulation. This formulation requires a once-monthly injection.


Naltrexone is “contraindicated” under the following conditions:



  • (1)

    Patients with hypersensitivity reactions to naltrexone.


  • (2)

    Patients who have previously exhibited hypersensitivity to naltrexone, polylactide-coglycolide, carboxymethyl-cellulose, or any other components of the diluent (for extended-release injectable naltrexone).


  • (3)

    Patients with current physical dependence on opioids, including partial agonists.


  • (4)

    Patients with current physiologic opioid dependence.


  • (5)

    Patients in acute opioid withdrawal.


  • (6)

    Any individual who has failed the naloxone challenge test (see “Glossary”) or has a positive urine screen for opioids.



Naltrexone should be used with “caution” under the following conditions:



  • (1)

    All patients should be warned of the risk of hepatic injury and advised to seek medical attention if they experience symptoms of acute hepatitis. Hepatic injury is a concern if very high doses are used, for example, 200–300 mg per day. Use of naltrexone should be discontinued in the event of symptoms and/or signs of acute hepatitis. Cases of hepatitis and clinically significant liver dysfunction were observed in association with naltrexone exposure during the clinical development program and in the postmarketing period. Transient, asymptomatic hepatic transaminase elevations were also observed in the clinical trials and postmarketing period.


  • (2)

    Patients with liver impairment should complete liver enzyme tests before and during treatment with naltrexone to check for additional liver impairment.


  • (3)

    Patients who experience injection site reactions should be monitored for pain, redness, or swelling. Incorrect administration may increase the risk of injection site reactions. Reactions have occurred with extended-release injectable naltrexone.


  • (4)

    Patients with co-occurring psychiatric disorders should be monitored for adverse events. Suicidal thoughts, attempted suicide, and depression have been reported.



Significant “medication interactions” with naltrexone are as follows:



  • (1)

    Naltrexone should not be used with methylnaltrexone or naloxegol.


  • (2)

    Naltrexone blocks the effects of opioid analgesics because it is an opioid antagonist.


  • (3)

    Glyburide may increase serum concentration of naltrexone. Monitor for increased toxicity effects of naltrexone.



Summary of Recommendations




  • (1)

    The choice of available treatment options for addiction involving opioid use should be a shared decision between the clinician and the patient.


  • (2)

    Clinicians should consider the patient’s preferences, past treatment history, and treatment setting when deciding between the use of methadone, buprenorphine, and naltrexone in the treatment of addiction involving opioid use.



The treatment setting described as level 1 treatment in the ASAM Criteria may be a general outpatient location such as a clinician’s practice site. The setting as described as level 2 in the ASAM Criteria may be an intensive outpatient treatment or partial hospitalization program housed in a specialty addiction treatment facility, a community mental health center, or another setting.


The ASAM Criteria describes level 3 or level 4 treatment, respectively, as a residential addiction treatment facility or hospital.



  • (1)

    The venue in which treatment is provided is as important as the specific medication selected. OTPs offer daily supervised dosing of methadone, and increasingly of buprenorphine. In accordance with Federal law (21 CFR §1306.07), OBOT, which provides medication on a prescribed weekly or monthly basis, is limited to buprenorphine. 9 Naltrexone can be prescribed in any setting by any clinician with the authority to prescribe any medication. Clinicians should consider a patient’s psychosocial situation, co-occurring disorders, and risk of diversion when determining whether OTP or OBOT is most appropriate.


  • (2)

    OBOT may not be suitable for patients with active alcohol use disorder or sedative, hypnotic, or anxiolytic use disorder (or who are in the treatment of addiction involving the use of alcohol or other sedative drugs, including benzodiazepines or benzodiazepine receptor agonists). It may also be unsuitable for persons who are regularly using alcohol or other sedatives, but do not have addiction or a specific substance use disorder related to that class of drugs. The prescribing of benzodiazepines or other sedative-hypnotics should be used with extreme caution in patients who are prescribed methadone or buprenorphine for the treatment of an opioid use disorder.


  • (3)

    Methadone is recommended for patients who may benefit from daily dosing and supervision in an OTP, or for patients for whom buprenorphine for the treatment of opioid use disorder has been used unsuccessfully in an OTP or OBOT setting.


  • (4)

    Oral naltrexone for the treatment of opioid use disorder is often adversely affected by poor medication adherence. Clinicians should reserve its use for patients who would be able to comply with special techniques to enhance their adherence, for example, observed dosing. Extended release injectable naltrexone reduces, but does not eliminate, issues with medication adherence.



Areas for Further Research


More research is needed to compare the advantages of agonists and antagonists in the treatment of opioid use disorder. Whereas methadone, buprenorphine, and naltrexone are all superior to no treatment in opioid use disorder, less is known about their relative advantages.

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Jan 19, 2020 | Posted by in PATHOLOGY & LABORATORY MEDICINE | Comments Off on The ASAM National Practice Guideline for the Use of Medications in the Treatment of Addiction Involving Opioid Use ∗

Full access? Get Clinical Tree

Get Clinical Tree app for offline access