Opioid use disorder

Last updated

Opioid use disorder
Other namesOpioid addiction, [1] problematic opioid use, [1] opioid abuse, [2] opioid dependence [3]
Morphin - Morphine.svg
Molecular structure of morphine
Specialty Addiction medicine, psychiatry
Symptoms Strong desire to use opioids, increased tolerance to opioids, failure to meet obligations, trouble with reducing use, withdrawal syndrome with discontinuation [4] [5]
Complications Opioid overdose, hepatitis C, marriage problems, unemployment, poverty [4] [5]
DurationLong term [6]
Causes Opioids [3]
Diagnostic method Based on criteria in the DSM-5 [4]
Differential diagnosis Alcoholism
Treatment Opioid replacement therapy, behavioral therapy, twelve-step programs, take home naloxone [7] [8] [9]
Medication Buprenorphine, methadone, naltrexone [7] [10]
Frequency16 million [11]
Deaths120,000 [11]

Opioid use disorder (OUD) is a substance use disorder characterized by cravings for opioids, continued use despite physical and/or psychological deterioration, increased tolerance with use, and withdrawal symptoms after discontinuing opioids. [12] Opioid withdrawal symptoms include nausea, muscle aches, diarrhea, trouble sleeping, agitation, and a low mood. [5] Addiction and dependence are important components of opioid use disorder. [13]

Contents

Risk factors include a history of opioid misuse, current opioid misuse, young age, socioeconomic status, race, untreated psychiatric disorders, and environments that promote misuse (social, family, professional, etc.). [14] [15] Complications may include opioid overdose, suicide, HIV/AIDS, hepatitis C, and problems meeting social or professional responsibilities. [5] [4] Diagnosis may be based on criteria by the American Psychiatric Association in the DSM-5. [4]

Opioids include substances such as heroin, morphine, fentanyl, codeine, dihydrocodeine, oxycodone, and hydrocodone. [5] [6] A useful standard for the relative strength of different opioids is morphine milligram equivalents (MME). [16] It is recommended for clinicians to refer to daily MMEs when prescribing opioids to decrease the risk of misuse and adverse effects. [17]

Long-term opioid use occurs in about 4% of people following their use for trauma or surgery-related pain. [18] In the United States, most heroin users begin by using prescription opioids that may also be bought illegally. [19] [20]

People with an opioid use disorder are often treated with opioid replacement therapy using methadone or buprenorphine. [21] Such treatment reduces the risk of death. [21] Additionally, they may benefit from cognitive behavioral therapy, other forms of support from mental health professionals such as individual or group therapy, twelve-step programs, and other peer support programs. [22] The medication naltrexone may also be useful to prevent relapse. [10] [8] Naloxone is useful for treating an opioid overdose and giving those at risk naloxone to take home is beneficial. [23] In 2020, the CDC estimated that nearly 3 million people in the U.S. were living with OUD and more than 65,000 people died by opioid overdose, of whom more than 15,000 were heroin overdoses. [24] [25]

Diagnosis

The DSM-5 guidelines for the diagnosis of opioid use disorder require that the individual has a significant impairment or distress related to opioid uses. [4] To make the diagnosis two or more of 11 criteria must be present in a given year: [4]

  1. More opioids are taken than intended
  2. The individual is unable to decrease the number of opioids used
  3. Large amounts of time are spent trying to obtain opioids, use opioids, or recover from taking them
  4. The individual has cravings for opioids
  5. Difficulty fulfilling professional duties at work or school
  6. Continued use of opioids leading to social and interpersonal consequences
  7. Decreased social or recreational activities
  8. Using opioids despite being in physically dangerous settings
  9. Continued use despite opioids worsening physical or psychological health (i.e. depression, constipation)
  10. Tolerance
  11. Withdrawal

The severity can be classified as mild, moderate, or severe based on the number of criteria present. [6] The tolerance and withdrawal criteria are not considered to be met for individuals taking opioids solely under appropriate medical supervision. [4] Addiction and dependence are components of a substance use disorder; addiction is the more severe form. [13]

Signs and symptoms

Opioid intoxication

Signs and symptoms of opioid intoxication include: [5] [26]

Opioid overdose

Fentanyl. 2 mg (white powder to the right) is a lethal dose in most people. US penny is 19 mm (0.75 in) wide. Fentanyl. 2 mg. A lethal dose in most people.jpg
Fentanyl. 2 mg (white powder to the right) is a lethal dose in most people. US penny is 19 mm (0.75 in) wide.

Signs and symptoms of opioid overdose include, but are not limited to: [28]

Withdrawal

Opioid withdrawal can occur with a sudden decrease in, or cessation of, opioids after prolonged use. [29] [30] Onset of withdrawal depends on the half-life of the opioid that was used last. [31] With heroin this typically occurs five hours after use; with methadone, it may take two days. [31] The length of time that major symptoms occur also depends on the opioid used. [31] For heroin withdrawal, symptoms are typically greatest at two to four days and can last up to two weeks. [32] [31] Less significant symptoms may remain for an even longer period, in which case the withdrawal is known as post-acute-withdrawal syndrome. [31]

Treatment of withdrawal may include methadone and buprenorphine. Medications for nausea or diarrhea may also be used. [30]

Cause

Opioid use disorder can develop as a result of self-medication. [33] Scoring systems have been derived to assess the likelihood of opiate addiction in chronic pain patients. [34] Healthcare practitioners have long been aware that despite the effective use of opioids for managing pain, empirical evidence supporting long-term opioid use is minimal. [35] [36] [37] [38] [39] Many studies of patients with chronic pain have failed to show any sustained improvement in their pain or ability to function with long-term opioid use. [36] [40] [41] [42] [39]

According to position papers on the treatment of opioid dependence published by the United Nations Office on Drugs and Crime and the World Health Organization, care providers should not treat opioid use disorder as the result of a weak moral character or will but as a medical condition. [15] [43] [44] Some evidence suggests the possibility that opioid use disorders occur due to genetic or other chemical mechanisms that may be difficult to identify or change, such as dysregulation of brain circuitry involving reward and volition. But the exact mechanisms involved are unclear, leading to debate over the influence of biology and free will. [45] [46]

Mechanism

Addiction

Addiction is a brain disorder characterized by compulsive drug use despite adverse consequences. [13] [47] [48] [49] Addiction involves the overstimulation of the brain's mesocorticolimbic reward circuit (reward system), essential for motivating behaviors linked to survival and reproductive fitness, like seeking food and sex. [50] This reward system encourages associative learning and goal-directed behavior. In addiction, substances overactivate this circuit, causing compulsive behavior due to changes in brain synapses. [51]

The incentive-sensitization theory differentiates between "wanting" (driven by dopamine in the reward circuit) and "liking" (related to brain pleasure centers). [52] This explains the addictive potential of non-pleasurable substances and the persistence of opioid addiction despite tolerance to their euphoric effects. Addiction surpasses mere avoidance of withdrawal, involving cues and stress that reactivate reward-driven behaviors. [50] This is an important reason detoxification alone is unsuccessful 90% of the time. [53] [54] [55]

Mesocorticolimbic circuit which plays major role in addiction that is affected by opioids Mesocorticolimbic Circuit.png
Mesocorticolimbic circuit which plays major role in addiction that is affected by opioids

Overexpression of the gene transcription factor ΔFosB in the nucleus accumbens plays a crucial role in the development of an addiction to opioids and other addictive drugs by sensitizing drug reward and amplifying compulsive drug-seeking behavior. [47] [56] [57] [58] Like other addictive drugs, overuse of opioids leads to increased ΔFosB expression in the nucleus accumbens. [56] [57] [58] [59] Opioids affect dopamine neurotransmission in the nucleus accumbens via the disinhibition of dopaminergic pathways as a result of inhibiting the GABA-based projections to the ventral tegmental area (VTA) from the rostromedial tegmental nucleus (RMTg), which negatively modulate dopamine neurotransmission. [60] [61] In other words, opioids inhibit the projections from the RMTg to the VTA, which in turn disinhibits the dopaminergic pathways that project from the VTA to the nucleus accumbens and elsewhere in the brain. [60] [61]

The differences in the genetic regions encoding the dopamine receptors for each individual may help to elucidate part of the risk for opioid addiction and general substance abuse. Studies of the D2 Dopamine Receptor, in particular, have shown some promising results. One specific SNP is at the TaqI RFLP (rs1800497). In a study of 530 Han Chinese heroin-addicted individuals from a Methadone Maintenance Treatment Program, those with the specific genetic variation showed higher mean heroin consumption by around double those without the SNP. [62] This study helps to show the contribution of dopamine receptors to substance addiction and more specifically to opioid abuse. [62]

Neuroimaging has shown functional and structural alterations in the brain. [63] Chronic intake of opioids such as heroin may cause long-term effects in the orbitofrontal area (OFC), which is essential for regulating reward-related behaviors, emotional responses, and anxiety. [64] Moreover, neuroimaging and neuropsychological studies demonstrate dysregulation of circuits associated with emotion, stress and high impulsivity. [65]

Dependence

Opioid dependence can occur as physical dependence, psychological dependence, or both. [66] Drug dependence is an adaptive state associated with a withdrawal syndrome upon cessation of repeated exposure to a stimulus (e.g., drug intake). [47] [48] [49] Dependence is a component of a substance use disorder. [13] [67] Opioid dependence can manifest as physical dependence, psychological dependence, or both. [66] [48] [67]

Increased brain-derived neurotrophic factor (BDNF) signaling in the ventral tegmental area (VTA) has been shown to mediate opioid-induced withdrawal symptoms via downregulation of insulin receptor substrate 2 (IRS2), protein kinase B (AKT), and mechanistic target of rapamycin complex 2 (mTORC2). [47] [68] As a result of downregulated signaling through these proteins, opiates cause VTA neuronal hyperexcitability and shrinkage (specifically, the size of the neuronal soma is reduced). [47] It has been shown that when an opiate-naive person begins using opiates in concentrations that induce euphoria, BDNF signaling increases in the VTA. [69]

Upregulation of the cyclic adenosine monophosphate (cAMP) signal transduction pathway by cAMP response element binding protein (CREB), a gene transcription factor, in the nucleus accumbens is a common mechanism of psychological dependence among several classes of drugs of abuse. [66] [47] Upregulation of the same pathway in the locus coeruleus is also a mechanism responsible for certain aspects of opioid-induced physical dependence. [66] [47]

A scale was developed to compare the harm and dependence liability of 20 drugs. [70] The scale uses a rating of zero to three to rate physical dependence, psychological dependence, and pleasure to create a mean score for dependence. [70] Selected results can be seen in the chart below. Heroin and morphine both scored highest, at 3.0. [70]

DrugMeanPleasurePsychological dependencePhysical dependence
Heroin/Morphine 3.003.03.03.0
Cocaine 2.393.02.81.3
Alcohol 1.932.31.91.6
Benzodiazepines 1.831.72.11.8
Tobacco 2.212.32.61.8

Opioid receptors

A genetic basis for the efficacy of opioids in the treatment of pain has been demonstrated for several specific variations, but the evidence for clinical differences in opioid effects is ambiguous. [11] There is an estimated 50% genetic contribution to opioid use disorder. [11] [71] The pharmacogenomics of the opioid receptors and their endogenous ligands have been the subject of intensive activity in association studies. These studies test broadly for a number of phenotypes, including opioid dependence, cocaine dependence, alcohol dependence, methamphetamine dependence/psychosis, response to naltrexone treatment, personality traits, and others. Major and minor variants have been reported for every receptor and ligand coding gene in both coding sequences, as well as regulatory regions. [71] Research on endogenous opioid receptors has focused around OPRM1 gene, which encodes the μ-opioid receptor, and the OPRK1 and OPRD1 genes, which encode the κ and δ receptors, respectively. [71] Newer approaches shift away from analysis of specific genes and regions, and are based on an unbiased screen of genes across the entire genome, which have no apparent relationship to the phenotype in question. These GWAS studies yield a number of implicated genes, although many of them code for seemingly unrelated proteins in processes such as cell adhesion, transcriptional regulation, cell structure determination, and RNA, DNA, and protein handling/modifying. [72]

118A>G variant

While over 100 variants have been identified for the opioid mu-receptor, the most studied mu-receptor variant is the non-synonymous 118A>G variant, which results in functional changes to the receptor, including lower binding site availability, reduced mRNA levels, altered signal transduction, and increased affinity for beta-endorphin. In theory, all these functional changes would reduce the impact of exogenous opioids, requiring a higher dose to achieve the same therapeutic effect. This points to a potential for greater addictive capacity in individuals who require higher dosages to achieve pain control. But evidence linking the 118A>G variant to opioid dependence is mixed, with associations shown in a number of study groups, but negative results in other groups. One explanation for the mixed results is the possibility of other variants that are in linkage disequilibrium with the 118A>G variant and thus contribute to different haplotype patterns more specifically associated with opioid dependence. [73]

Non-opioid receptor genes

While opioid receptors have been the most widely studied, a number of other genes have been implicated in OUD. Higher numbers of (CA) repeats flanking the preproenkephalin gene, PENK, have been associated with opiate dependence. [74] There have been mixed results for the MCR2 gene, encoding melanocortin receptor type 2, implicating both protection and risk to heroin addiction. [74] A number of enzymes in the cytochrome P450 family may also play a role in dependence and overdose due to variance in breakdown of opioids and their receptors. There are also multiple potential complications with combining opioids with antidepressants and antiepileptic drugs (both common drugs for chronic pain patients) because of their effects on inducing CYP enzymes. [75] Genotyping of CYP2D6 in particular may play a role in helping patients with individualized treatment for OUD and other drug addictions. [75]

Prevention

The CDC gives specific recommendations for prescribers regarding initiation of opioids, clinically appropriate use of opioids, and assessing possible risks associated with opioid therapy. [76] Large U.S. retail pharmacy chains are implementing protocols, guidelines, and initiatives to take back unused opioids, providing naloxone kits, and being vigilant for suspicious prescriptions. [77] [78] [79] Insurance programs can help limit opioid use by setting quantity limits on prescriptions or requiring prior authorizations for certain medications. [80]

Naloxone is used for the emergency treatment of an overdose. [81] It can be given by many routes (e.g., intramuscular (IM), intravenous (IV), subcutaneous, intranasal, and inhalation) and acts quickly by displacing opioids from opioid receptors and preventing the activation of these receptors. [79] Naloxone kits are recommended for laypersons who may witness an opioid overdose, for people with large prescriptions for opioids, those in substance use treatment programs, and those recently released from incarceration. [82] Since this is a life-saving medication, many areas of the U.S. have implemented standing orders for law enforcement to carry and give naloxone as needed. [83] [84] In addition, naloxone can be used to challenge a person's opioid abstinence status before starting a medication such as naltrexone, which is used in the management of opioid addiction. [85]

Good Samaritan laws typically protect bystanders who administer naloxone. In the U.S., at least 40 states have Good Samaritan laws to encourage bystanders to take action without fear of prosecution. [86] As of 2019, 48 states give pharmacists the authority to distribute naloxone without an individual prescription. [87]

Homicide, suicide, accidents and liver disease are also opioid-related causes of death for those with OUD. [88] [89] Many of these causes of death are unnoticed due to the often limited information on death certificates. [88] [90]

Mitigation

The "CDC Clinical Practice Guideline for Prescribing Opioids for Pain-United States, 2022" provides recommendations related to opioid misuse, OUD, and opioid overdoses. [16] It reports a lack of clinical evidence that "abuse-deterrent" opioids (e.g., OxyContin), as labeled by the U.S. Food and Drug Administration, are effective for OUD risk mitigation. [16] [91] CDC guidance suggests the prescription of immediate-release opioids instead of opioids that have a long duration (long-acting) or opioids that are released over time (extended release). [16] Other recommendations include prescribing the lowest opioid dose that successfully addresses the pain in opioid-naïve patients and collaborating with patients who already take opioid therapy to maximize the effect of non-opioid analgesics. [16]

While receiving opioid therapy, patients should be periodically evaluated for opioid-related complications and clinicians should review state prescription drug monitoring program systems. [16] The latter should be assessed to reduce the risk of overdoses in patients due to their opioid dose or medication combinations. [16] For patients receiving opioid therapy in whom the risks outweigh the benefits, clinicians and patients should develop a treatment plan to decrease their opioid dose incrementally. [16]

For more specific mitigation strategies regarding opioid overdoses, see opioid overdose § Prevention.

Management

Opioid use disorders typically require long-term treatment and care with the goal of reducing the person's risks and improving their long-term physical and psychological condition. [44]

First-line management involves the use of opioid replacement therapies, particularly methadone and buprenorphine/naloxone. Withdrawal management alone is strongly discouraged, because of its association with elevated risks of HIV and hepatitis C transmission, high rates of overdose deaths, and nearly universal relapse. [92] [93] This approach is seen as ineffective without plans for transition to long-term evidence-based addiction treatment, such as opioid agonist treatment. [53] Though treatment reduces mortality rates, the first four weeks after treatment begins and the four weeks after treatment ceases are the riskiest times for drug-related deaths. [7] These periods of increased vulnerability are significant because many of those in treatment leave programs during these periods. [7] There is evidence that people with opioid use disorder who are dependent on pharmaceutical opioids may require a different management approach from those who take heroin. [94]

Medication

Opioid replacement therapy (ORT), also known as opioid substitution therapy (OST) or Medications for Opioid Use Disorder (MOUD), involves replacing an opioid, such as heroin. [95] [96] Commonly used drugs for ORT are methadone and buprenorphine/naloxone (Suboxone), which are taken under medical supervision. [96] Buprenorphine/naloxone is usually preferred over methadone because of its safety profile, which is considered significantly better, primarily with regard to its risk of overdose [97] and effects on the heart (QTc prolongation). [98] [99]

Buprenorphine/naloxone, methadone, and naltrexone are approved by the U.S. Food and Drug Administration (FDA) for medication-assisted treatment (MAT). [100] In the U.S., the Substance Abuse and Mental Health Services Administration (SAMHSA) certifies opioid treatment programs (OTPs), where methadone can be dispensed at methadone clinics. [101] As of 2023, the Waiver Elimination (MAT Act), also known as the "Omnibus Bill", removed the federal requirement for medical providers to obtain a waiver to prescribe buprenorphine, in an attempt to increase access to OUD treatment. [102]

The driving principle behind ORT is the patient's reclamation of a self-directed life. [103] ORT facilitates this process by reducing symptoms of drug withdrawal and drug cravings. [96] [103] In some countries (not the U.S. or Australia), [96] regulations enforce a limited time for people on ORT programs that conclude when a stable economic and psychosocial situation is achieved. (People with HIV/AIDS or hepatitis C are usually excluded from this requirement.) In practice, 40–65% of patients maintain abstinence from additional opioids while receiving opioid replacement therapy and 70–95% can reduce their use significantly. [96] Medical (improper diluents, non-sterile injecting equipment), psychosocial (mental health, relationships), and legal (arrest and imprisonment) issues that can arise from the use of illegal opioids are concurrently eliminated or reduced. [96] Clonidine or lofexidine can help treat the symptoms of withdrawal. [104]

The period when initiating methadone and the time immediately after discontinuing treatment with both drugs are periods of particularly increased mortality risk, which should be dealt with by both public health and clinical strategies. [7] ORT has proved to be the most effective treatment for improving the health and living condition of people experiencing illegal opiate use or dependence, including mortality reduction [96] [105] [7] and overall societal costs, such as the economic loss from drug-related crime and healthcare expenditure. [96] A review of UK hospital policies found that local guidelines delayed access to substitute opioids, for instance by requiring lab tests to demonstrate recent use or input from specialist drug teams before prescribing. Delays to access can increase people's risk of discharging themselves early against medical advice. [106] [107] ORT is endorsed by the World Health Organization, United Nations Office on Drugs and Crime and UNAIDS as effective at reducing injection, lowering risk for HIV/AIDS, and promoting adherence to antiretroviral therapy. [7]

Buprenorphine and methadone work by reducing opioid cravings, easing withdrawal symptoms, and blocking the euphoric effects of opioids via cross-tolerance, [108] and in the case of buprenorphine, a high-affinity partial opioid agonist, also due to opioid receptor saturation. [109] It is this property of buprenorphine that can induce acute withdrawal when administered before other opioids with lower efficacy have left the body.

Buprenorphine and buprenorphine/naloxone

Buprenorphine can be administered either as a standalone product or in combination with the opioid antagonist naloxone. This inclusion is strategic: it deters misuse by preventing the crushing and injecting of the medication, encouraging instead the prescribed sublingual (under the tongue) route. [96] Buprenorphine/naloxone formulations are available as tablets and films; [110] these formulations operate efficiently when taken sublingually. In this form, buprenorphine's bioavailability remains robust (35–55%), while naloxone's is significantly reduced (~10%). [111] But when injected, naloxone's bioavailability increases, effectively blocking buprenorphine's opioid effects, thereby curbing potential abuse.

Buprenorphine's role as a partial opioid receptor agonist sets it apart from full agonists like methadone. Its unique pharmacological profile makes it less likely to cause respiratory depression, thanks to its "ceiling effect". [112] [113] While the risk of misuse or overdose is higher with buprenorphine alone compared to the buprenorphine/naloxone combination or methadone, its usage is linked to a decrease in mortality. [114] [7] Approved in the U.S. for opioid dependence treatment in 2002, [115] buprenorphine has since expanded in form, with the FDA approving a month-long injectable version in 2017. [116]

When initiating buprenorphine/naloxone therapy, several critical factors must be considered. These include the severity of withdrawal symptoms, the time elapsed since the last opioid use, and the type of opioid involved (long-acting vs. short-acting). [117] A standard induction method involves waiting until the patient exhibits moderate withdrawal symptoms, as measured by a Clinical Opiate Withdrawal Scale, achieving a score of around 12. Alternatively, "microdosing" commences with a small dose immediately, regardless of withdrawal symptoms, offering a more flexible approach to treatment initiation. [118] "Macrodosing" starts with a larger dose of Suboxone, a different induction strategy with its own set of considerations. [119]

Methadone

Methadone is a commonly used full-opioid agonist in the treatment of opioid use disorder. It is effective in relieving withdrawal symptoms and cravings in people with opioid addiction, and can also be used in pain control in certain situations. [114] While methadone is a widely prescribed form of OAT, it often requires more frequent clinical visits compared to buprenorphine/naloxone, which also has a better safety profile and lower risk of respiratory depression and overdose. [120]

Important considerations when initiating methadone include the patient's opioid tolerance, the time since last opioid use, the type of opioid used (long-acting vs. short-acting), and the risk of methadone toxicity. [121] Methadone comes in different forms: tablet, oral solution, or an injection. [114]

One of methadone's benefits is that it can last up to 56 hours in the body, so if a patient misses a daily dose, they will not typically struggle with withdrawal symptoms. [114] Other advantages of methadone include reduction in infectious disease related to injection drug use, and reduced mortality. Methadone has a number of potential side effects, including slowed breathing, nausea, vomiting, restlessness, and headache. [122]

Naltrexone

Naltrexone is an opioid receptor antagonist used for the treatment of opioid addiction. [123] [124] It is not as widely used as buprenorphine or methadone for OUD due to low rates of patient acceptance, non-adherence due to daily dosing, and difficulty achieving abstinence from opioids before beginning treatment. Additionally, dosing naltrexone after recent opioid use can lead to precipitated withdrawal. Conversely, naltrexone antagonism at the opioid receptor can be overcome with higher doses of opioids. [125] Naltrexone monthly IM injections received FDA approval in 2010 for the treatment of opioid dependence in abstinent opioid users. [123] [126]

Other opioids

Evidence of effects of heroin maintenance compared to methadone are unclear as of 2010. [127] A Cochrane review found some evidence in opioid users who had not improved with other treatments. [128] In Switzerland, Germany, the Netherlands, and the United Kingdom, long-term injecting drug users who do not benefit from methadone and other medication options may be treated with injectable heroin that is administered under the supervision of medical staff. [129] Other countries where it is available include Spain, Denmark, Belgium, Canada, and Luxembourg. [130] Dihydrocodeine in both extended-release and immediate-release form is also sometimes used for maintenance treatment as an alternative to methadone or buprenorphine in some European countries. [131] Dihydrocodeine is an opioid agonist. [132] It may be used as a second-line treatment. [133] A 2020 systematic review found low-quality evidence that dihydrocodeine may be no more effective than other routinely used medication interventions in reducing illicit opiate use. [134] An extended-release morphine confers a possible reduction of opioid use and with fewer depressive symptoms but overall more adverse effects compared to other forms of long-acting opioids. Retention in treatment was not found to be significantly different. [135] It is used in Switzerland and more recently in Canada. [136]

In pregnancy

Pregnant women with opioid use disorder can also receive treatment with methadone, naltrexone, or buprenorphine. [137] Buprenorphine appears to be associated with more favorable outcomes compared to methadone for treating opioid use disorder (OUD) in pregnancy. Studies show that buprenorphine is linked to lower risks of preterm birth, greater birth weight, and larger head circumference without increased harm. [138] Compared to methadone, it consistently results in improved birth weight and gestational age, though these findings should be interpreted with caution due to potential biases. [139] Buprenorphine use also correlates with a lower risk of adverse neonatal outcomes, with similar risks of adverse maternal outcomes as methadone. [140] Infants born to buprenorphine-treated mothers generally have higher birth weights, fewer withdrawal symptoms, and a lower likelihood of premature birth. [141] Additionally, these infants often require less treatment for neonatal abstinence syndrome and have mothers who are more likely to start treatment earlier in pregnancy, leading to longer gestations and larger infants. [142] These findings suggest buprenorphine may be a more favorable option for OUD treatment during pregnancy, but individual circumstances and treatment availability must be considered.

Behavioral therapy

Paralleling the variety of medical treatments, there are many forms of psychotherapy and community support for treating OUD. The primary evidence-based psychotherapies include cognitive behavioral therapy (CBT), motivational enhancement therapy (MET), contingency management (CM), and twelve-step programs. Community-based support such as support groups (e.g., Narcotics Anonymous) and therapeutic housing for those with OUD is also an important aspect of healing. [143] [144]

Cognitive behavioral therapy

Cognitive behavioral therapy (CBT) is a form of psychosocial intervention that systematically evaluates thoughts, feelings, and behaviors about a problem and works to develop coping strategies to work through those problems. [145] This intervention has demonstrated success in many psychiatric conditions (e.g., depression) and substance use disorders (e.g., tobacco). [146] But the use of CBT alone for OUD has declined due to lack of efficacy, and many rely on medication therapy or medication therapy with CBT, since both were found to be more efficacious than CBT alone. [147] CBT has been shown to be more successful in relapse prevention than treatment of ongoing drug use. [143] It is particularly known for its durability. [148]

Motivational Enhancement Therapy

Motivational enhancement therapy (MET) is the manualized form of motivational interviewing (MI). MI leverages one's intrinsic motivation to recover through education, formulation of relapse prevention strategies, reward for adherence to treatment guidelines, and positive thinking to keep motivation high—which are based on a person's socioeconomic status, gender, race, ethnicity, sexual orientation, and readiness to recover. [147] [149] [150] Like CBT, MET alone has not shown convincing efficacy for OUD. There is stronger support for combining it with other therapies. [148]

Contingency Management Therapy

Contingency Management Therapy (CMT) employs similar principles as operant behavioral conditioning, such as using incentives to reach certain goals (e.g., verified abstinence, usually in the form of urine drug testing). [143] This form of psychotherapy has the strongest, most robust empirical support for treating drug addiction. [143] [148] [151] Outpatient clients are shown to have improved medication compliance, retention, and abstinence when using voucher-based incentives. [143] [148] One way this is implemented is to offer take-home privileges for methadone programs. Despite its effectiveness during treatment, effects tend to wane once terminated. Additionally, the cost barrier limits its application in the clinical community. [143]

Twelve-step programs

While medical treatment may help with the initial symptoms of opioid withdrawal, once the first stages of withdrawal are through, a method for long-term preventative care is attendance at 12-step groups such as Narcotics Anonymous (NA). [152] NA's 12-step process is based on the 12-step facilitation of Alcoholic Anonymous (AA) and centers on peer support, self-help, and spiritual connectedness. Some evidence also supports the use of these programs for adolescents. [153] Multiple studies have shown increased abstinence for those in NA compared to those who are not. [11] [154] [155] [156] Members report a median abstinence length of 5 years. [156]

Novel experimental treatments

Though medications and behavioral treatments are effective forms for treating OUD, relapse remains a common problem. The medical community has looked to novel technologies and traditional alternative medicines for new ways to approach the issues of continued cravings and impaired executive functioning. While consensus on their efficacy has not been reached, a number of reviews have shown promising results for the use of non-invasive brain stimulation (NIBS) for reducing cravings in OUD. [157] [158] These results are consistent with the use of NIBS for reducing cravings of other substances. Additionally, investigations into the anecdotal evidence of psychedelics like ibogaine have also shown the possibility of decreased cravings and withdrawal symptoms. [159] Ibogaine is illegal in the U.S. but is unregulated in Mexico, Costa Rica, and New Zealand, where many clinics use it for addiction treatment. [160] Research has shown a minor mortality risk due to its cardiotoxic and neurotoxic effects. [159]

Epidemiology

A two milligram dose of fentanyl powder (on pencil tip) is a lethal amount for most people. 2 milligrams of fentanyl on pencil tip. A lethal dose for most people. US Drug Enforcement Administration.jpg
A two  milligram dose of fentanyl powder (on pencil tip) is a lethal amount for most people.

Globally, the number of people with opioid dependence increased from 10.4 million in 1990 to 15.5 million in 2010. [7] In 2016, the numbers rose to 27 million people who experienced this disorder. [162] Opioid use disorders resulted in 122,000 deaths worldwide in 2015, [163] up from 18,000 deaths in 1990. [164] Deaths from all causes rose from 47.5 million in 1990 to 55.8 million in 2013. [164] [163]

United States

3 waves of opioid overdose deaths in the U.S. 3 waves of opioid overdose deaths. US timeline.png
3 waves of opioid overdose deaths in the U.S.

The current epidemic of opioid abuse is the most lethal drug epidemic in American history. [20] The crisis can be distinguished by waves of opioid overdose deaths as described by the Centers of Disease Control and Prevention. [165] The first wave began in the 1990s, related to the rise in prescriptions of natural opioids (such as codeine and morphine), semisynthetic opioids (oxycodone, hydrocodone, hydromorphone, and oxymorphone), and synthetic opioids like methadone. [166] [165] In the U.S., "the age-adjusted drug poisoning death rate involving opioid analgesics increased from 1.4 to 5.4 deaths per 100,000 population between 1999 and 2010. [167] The second wave dates to around 2010 with the rapid increase in opioid overdoses due to heroin. [166] By this time, there were already four times as many deaths by overdose than in 1999. [167] The age-adjusted drug poisoning death rate involving heroin doubled from 0.7 to 1.4 deaths per 100,000 people between 1999 and 2011 and then continued to increase to 4.1 in 2015. [168] The third wave of overdose deaths began in 2013, related to synthetic opioids, particularly illicitly produced fentanyl. [166] While the illicit fentanyl market has continuously changed, the drug is generally sold as an adulterant in heroin. Research suggests that the rapid increase of fentanyl into the illicit opioid market has been largely supply-side-driven and dates to 2006. Decreasing heroin purity, competition from increased access to prescription medications, and dissemination of "The Siegfried Method" (a relatively simple and cost-effective method of fentanyl production) were major factors in street suppliers' inclusion of fentanyl in their products. [169] [170] The current, fourth wave, which began in 2016, has been characterized by polysubstance overdose due to synthetic opioids like fentanyl mixed with stimulants such as methamphetamine or cocaine. [171] [172] In 2010, around 0.5% of opioid-related deaths were attributed to mixture with stimulants. This figure increased more than 50-fold by 2021, when about a third of opioid-related deaths, or 34,000, involved stimulant use. [172]

In 2017, the U.S. Department of Health and Human Services (HHS) announced a public health emergency due to an increase in the misuse of opioids. [173] The administration introduced a strategic framework called the Five-Point Opioid Strategy, which includes providing access recovery services, increasing the availability of reversing agents for overdose, funding opioid misuse and pain research, changing treatments of people managing pain, and updating public health reports related to combating opioid drug misuse. [173] [174]

The U.S. epidemic in the 2000s is related to a number of factors. [15] Rates of opioid use and dependency vary by age, sex, race, and socioeconomic status. [15] With respect to race, the discrepancy in deaths is thought to be due to an interplay between physician prescribing and lack of access to healthcare and certain prescription drugs. [15] Men are at higher risk for opioid use and dependency than women, [175] [176] and men also account for more opioid overdoses than women, although this gap is closing. [175] Women are more likely to be prescribed pain relievers, be given higher doses, use them for longer durations, and become dependent upon them faster. [177]

Deaths due to opioid use also tend to skew at older ages than deaths from use of other illicit drugs. [176] [178] [179] This does not reflect opioid use as a whole, which includes younger people. Overdoses from opioids are highest among people between the ages of 40 and 50, [179] in contrast to heroin overdoses, which are highest among people between the ages of 20 and 30. [178] 21- to 35-year-olds represent 77% of people who enter treatment for opioid use disorder, [180] but the average age of first-time use of prescription painkillers was 21.2 years in 2013. [181] Among the middle class, means of acquiring funds include elder financial abuse and international dealers noticing a lack of enforcement in their transaction scams throughout the Caribbean. [182]

Since 2018, with the federal government's passing of the SUPPORT (Substance Use-Disorder Prevention That Promotes Opioid Recovery and Treatment for Patients and Communities Act) Act, federal restrictions on methadone use for patients receiving Medicare have been lifted. [183] Since March 2020, as a result of the COVID-19 pandemic, buprenorphine may be dispensed via telemedicine in the U.S. [184] [185]

In October 2021, New York Governor Kathy Hochul signed legislation to combat the opioid crisis. This included establishing a program for the use of medication-assisted substance use disorder treatment for incarcerated individuals in state and local correctional facilities, decriminalizing the possession and sale of hypodermic needles and syringes, establishing an online directory for distributors of opioid antagonists, and expanding the number of eligible crimes committed by individuals with a substance use disorder that may be considered for diversion to a substance use treatment program. [186] Until these laws were signed, incarcerated New Yorkers did not reliably have access to medication-assisted treatment and syringe possession was still a class A misdemeanor despite New York authorizing and funding syringe exchange and access programs. [187] This legislation acknowledges the ways New York State laws have contributed to opioid deaths: in 2020 more than 5112 people died from overdoses in New York State, with 2192 deaths in New York City. [188]

As of 2023, the Waiver Elimination (MAT Act), as part of Section 1262 of the Consolidated Appropriations Act, 2023 (or "Omnibus Bill"), removed the federal requirement for medical providers to obtain a waiver to prescribe buprenorphine, in an attempt to increase access to OUD treatment. [102] Before this bill, practitioners were required to receive a Drug Addiction Treatment Act of 2000 (DATA) waiver, also known as "x-waiver", before prescribing buprenorphine. There is also now no longer any limit to the number of patients to whom a provider may prescribe buprenorphine for OUD. [102]

Effects of COVID-19 on opioid overdose and telehealth treatment

Epidemiological research has shown that the COVID-19 pandemic accelerated the opioid crisis. [170] [191] [192] The overarching trend of opioid overdose data has shown a plateau in deaths around 2017–18, with a sudden and acute rise in 2019 primarily attributed to synthetic opioids like fentanyl. [190] In 2020, there were 93,400 drug overdoses in the U.S. with >73% (approximately 69,000) due to opioid overdose. [193] One JAMA review by Gomes et al. showed that estimated years of life loss (YLL) due to opioid toxicity in the U.S. increased by 276%. This increase was particularly felt by those ages 15 to 19, whose YLL increased nearly threefold. Younger male adults had the largest effect size. [192] Other reviews of U.S. and Canadian opioid data coinciding with the onset of COVID-19 suggested significant increases in opioid-related emergency medicine utilization, increased positivity for opioids, and surprisingly no to decreased change in naloxone dispensation. [194]

Telehealth played a large role in OUD treatment access, and legislation on telehealth continues to evolve. A study of Medicare beneficiaries with new-onset OUD showed that those who received telehealth services had a 33% lower risk of death by overdose. [195] Minority groups such as Black and Hispanic Americans have also been shown to benefit from the increased access due to telehealth programs introduced during the pandemic, despite increasing disparity gaps in other OUD-related outcomes. [196] The DEA and HHS have extended telemedicine flexibility in regard to prescribing controlled substances such as buprenorphine for OUD through December 31, 2024. [197]

History

Opiate misuse has been recorded at least since 300 BC. Greek mythology describes Nepenthe (Greek "free from sorrow") and how it was used by the hero of the Odyssey. Opioids have been used in the Near East for centuries. The purification of and isolation of opiates occurred in the early 19th century. [28]

Levacetylmethadol was previously used to treat opioid dependence. In 2003 the drug's manufacturer discontinued production. There are no available generic versions. LAAM produced long-lasting effects, which allowed the person receiving treatment to visit a clinic only three times per week, as opposed to daily as with methadone. [198] In 2001, levacetylmethadol was removed from the European market due to reports of life-threatening ventricular rhythm disorders. [199] In 2003, Roxane Laboratories, Inc. discontinued Orlaam in the U.S. [200]

See also

Related Research Articles

<span class="mw-page-title-main">Methadone</span> Opioid medication

Methadone, sold under the brand names Dolophine and Methadose among others, is a synthetic opioid agonist used for chronic pain and also for opioid use disorder. It is used to treat chronic pain, and it is also used to treat addiction to heroin or other opioids. Prescribed for daily use, the medicine relieves cravings and removes withdrawal symptoms. Withdrawal management using methadone can be accomplished in less than a month, or it may be done gradually over a longer period of time, or simply maintained for the rest of the patient's life. While a single dose has a rapid effect, maximum effect can take up to five days of use. After long-term use, in people with normal liver function, effects last 8 to 36 hours. Methadone is usually taken by mouth and rarely by injection into a muscle or vein.

<span class="mw-page-title-main">Harm reduction</span> Public health policies which lessen negative aspects of problematic activities

Harm reduction, or harm minimization, refers to a range of intentional practices and public health policies designed to lessen the negative social and/or physical consequences associated with various human behaviors, both legal and illegal. Harm reduction is used to decrease negative consequences of recreational drug use and sexual activity without requiring abstinence, recognizing that those unable or unwilling to stop can still make positive change to protect themselves and others.

<span class="mw-page-title-main">Naloxone</span> Opioid receptor antagonist

Naloxone is an opioid antagonist: a medication used to reverse or reduce the effects of opioids. For example, it is used to restore breathing after an opioid overdose. Effects begin within two minutes when given intravenously, five minutes when injected into a muscle, and ten minutes as a nasal spray. Naloxone blocks the effects of opioids for 30 to 90 minutes.

<span class="mw-page-title-main">Buprenorphine</span> Opioid used to treat pain & opioid use disorder

Buprenorphine, sold under the brand name Subutex among others, is an opioid used to treat opioid use disorder, acute pain, and chronic pain. It can be used under the tongue (sublingual), in the cheek (buccal), by injection, as a skin patch (transdermal), or as an implant. For opioid use disorder, the patient must have moderate opioid withdrawal symptoms before buprenorphine can be administered under direct observation of a health-care provider.

<span class="mw-page-title-main">Naltrexone</span> Medication

Naltrexone, sold under the brand name Revia among others, is a medication primarily used to manage alcohol use or opioid use disorder by reducing cravings and feelings of euphoria associated with substance use disorder. It has also been found effective in the treatment of other addictions and may be used for them off-label. An opioid-dependent person should not receive naltrexone before detoxification. It is taken by mouth or by injection into a muscle. Effects begin within 30 minutes, though a decreased desire for opioids may take a few weeks to occur. Side effects may include trouble sleeping, anxiety, nausea, and headaches. In those still on opioids, opioid withdrawal may occur. Use is not recommended in people with liver failure. It is unclear if use is safe during pregnancy. Naltrexone is an opioid antagonist and works by blocking the effects of opioids, including both opioid drugs as well as opioids naturally produced in the brain.

Substance dependence, also known as drug dependence, is a biopsychological situation whereby an individual's functionality is dependent on the necessitated re-consumption of a psychoactive substance because of an adaptive state that has developed within the individual from psychoactive substance consumption that results in the experience of withdrawal and that necessitates the re-consumption of the drug. A drug addiction, a distinct concept from substance dependence, is defined as compulsive, out-of-control drug use, despite negative consequences. An addictive drug is a drug which is both rewarding and reinforcing. ΔFosB, a gene transcription factor, is now known to be a critical component and common factor in the development of virtually all forms of behavioral and drug addictions, but not dependence.

A methadone clinic is a medical facility where medications for opioid use disorder (MOUD) are dispensed-—historically and most commonly methadone, although buprenorphine is also increasingly prescribed. Medically assisted drug therapy treatment is indicated in patients who are opioid-dependent or have a history of opioid dependence. Methadone is a schedule II (USA) opioid analgesic, that is also prescribed for pain management. It is a long-acting opioid that can delay the opioid withdrawal symptoms that patients experience from taking short-acting opioids, like heroin, and allow time for withdrawal management. In the United States, by law, patients must receive methadone under the supervision of a physician, and dispensed through an Opioid Treatment Program (OTP) certified by the Substance Abuse and Mental Health Services Administration and registered with the Drug Enforcement Administration.

<span class="mw-page-title-main">Opioid antagonist</span> Receptor agonist that acts on one or more of the opioid receptors

An opioid antagonist, or opioid receptor antagonist, is a receptor antagonist that acts on one or more of the opioid receptors.

Drug detoxification is variously construed or interpreted as a type of "medical" intervention or technique in regards to a physical dependence mediated by a drug; as well as the process and experience of a withdrawal syndrome or any of the treatments for acute drug overdose (toxidrome). The first definition however, in relation to substance dependence and its treatment is arguably a misnomer and even directly contradictory since withdrawal is neither contingent upon nor alleviated through biological excretion or clearance of the drug. In fact, excretion of a given drug from the body is one of the very processes that leads to withdrawal since the syndrome arises largely due to the cessation itself and the drug being absent from the body; especially the blood plasma, not from ‘leftover toxins’ or traces of the drug still being in the system.

<span class="mw-page-title-main">Opioid overdose</span> Toxicity due to excessive consumption of opioids

An opioid overdose is toxicity due to excessive consumption of opioids, such as morphine, codeine, heroin, fentanyl, tramadol, and methadone. This preventable pathology can be fatal if it leads to respiratory depression, a lethal condition that can cause hypoxia from slow and shallow breathing. Other symptoms include small pupils and unconsciousness; however, its onset can depend on the method of ingestion, the dosage and individual risk factors. Although there were over 110,000 deaths in 2017 due to opioids, individuals who survived also faced adverse complications, including permanent brain damage.

<span class="mw-page-title-main">Substance use disorder</span> Continual use of drugs (including alcohol) despite detrimental consequences

Substance use disorder (SUD) is the persistent use of drugs despite the substantial harm and adverse consequences to one's own self and others, as a result of their use. In perspective, the effects of the wrong use of substances that are capable of causing harm to the user or others, have been extensively described in different studies using a variety of terms such as substance use problems, problematic drugs or alcohol use, and substance use disorder. The National Institute of Mental Health (NIMH) states that "Substance use disorder (SUD) is a treatable mental disorder that affects a person's brain and behavior, leading to their inability to control their use of substances like legal or illegal drugs, alcohol, or medications. Symptoms can be moderate to severe, with addiction being the most severe form of SUD". Substance use disorders (SUD) are considered to be a serious mental illness that fluctuates with the age that symptoms first start appearing in an individual, the time during which it exists and the type of substance that is used. It is not uncommon for those who have SUD to also have other mental health disorders. Substance use disorders are characterized by an array of mental/emotional, physical, and behavioral problems such as chronic guilt; an inability to reduce or stop consuming the substance(s) despite repeated attempts; operating vehicles while intoxicated; and physiological withdrawal symptoms. Drug classes that are commonly involved in SUD include: alcohol (alcoholism); cannabis; opioids; stimulants such as nicotine, cocaine and amphetamines; benzodiazepines; barbiturates; and other substances.

Neonatal withdrawal or neonatal abstinence syndrome (NAS) or neonatal opioid withdrawal syndrome (NOWS) is a withdrawal syndrome of infants after birth caused by in utero exposure to drugs of dependence, most commonly opioids. Common signs and symptoms include tremors, irritability, vomiting, diarrhea, and fever. NAS is primarily diagnosed with a detailed medication history and scoring systems. First-line treatment should begin with non-medication interventions to support neonate growth, though medication interventions may be used in certain situations.

<span class="mw-page-title-main">Buprenorphine/naloxone</span> Opioid treatment

Buprenorphine/naloxone, sold under the brand name Suboxone among others, is a fixed-dose combination medication that includes buprenorphine and naloxone. It is used to treat opioid use disorder, and reduces the mortality of opioid use disorder by 50%. It relieves cravings to use and withdrawal symptoms. Buprenorphine/­naloxone is available for use in two different forms, under the tongue or in the cheek.

<span class="mw-page-title-main">Opioid epidemic in the United States</span> Ongoing overuse of opioid medication in the US

There is an ongoing opioid epidemic in the United States, originating out of both medical prescriptions and illegal sources. The epidemic began in the United States in the late 1990s, according to the Centers for Disease Control and Prevention (CDC), when opioids were increasingly prescribed for pain management, resulting in a rise in overall opioid use throughout subsequent years.

<span class="mw-page-title-main">Opioid epidemic</span> Deaths due to abuse of opioid drugs

The opioid epidemic, also referred to as the opioid crisis, is the rapid increase in the overuse, misuse/abuse, and overdose deaths attributed either in part or in whole to the class of drugs called opiates/opioids since the 1990s. It includes the significant medical, social, psychological, demographic and economic consequences of the medical, non-medical, and recreational abuse of these medications.

Clinical Opiate Withdrawal Scale (COWS) is a method used by registered practitioners to measure the severity of a patient's opioid withdrawal symptoms. This method consists of a series of 11 topics each comprising 4 - 5 common symptoms experienced by a patient undergoing opioid withdrawal. In each topic a rank is given depending on what the patient responds to. Generally, 0 is considered to be no symptom shown and 4 or 5 is considered to be the most common and severe symptom shown. These results are then added up and a final diagnosis is made based on the value obtained. This test is crucial as it allows the practitioner to assess the physiological and psychological behaviours of the patient as well as the severity of each symptom during the duration of the examination. The results are grouped into 3 categories of mild, moderately severe and severe. Mild consists of 5 to 12 points, moderately severe consists of 13 to 24 points and anything above 36 points is severe and requires direct medical attention.

<span class="mw-page-title-main">Opioid withdrawal</span> Withdrawal symptoms of opiates

Opioid withdrawal is a set of symptoms arising from the sudden withdrawal or reduction of opioids where previous usage has been heavy and prolonged. Signs and symptoms of withdrawal can include drug craving, anxiety, restless legs, nausea, vomiting, diarrhea, sweating, and an increased heart rate. Opioid use triggers a rapid adaptation in cellular signalling pathways that means, when rapidly withdrawn, there can be adverse physiological effects. All opioids, both recreational drugs and medications, when reduced or stopped, can lead to opioid withdrawal symptoms. When withdrawal symptoms are due to recreational opioid use, the term opioid use disorder is used, whereas when due to prescribed medications, the term prescription opioid use disorder is used. Opioid withdrawal can be helped by the use of opioid replacement therapy, and symptoms may be relieved by the use of medications including lofexidine and clonidine.

Opioid agonist therapy (OAT) is a treatment in which prescribed opioid agonists are given to patients who live with Opioid use disorder (OUD). In the case of methadone maintenance treatment (MMT), methadone is used to treat dependence on heroin or other opioids, and is administered on an ongoing basis.

<span class="mw-page-title-main">Prescription drug addiction</span> Medical condition

Prescription drug addiction is the chronic, repeated use of a prescription drug in ways other than prescribed for, including using someone else’s prescription. A prescription drug is a pharmaceutical drug that may not be dispensed without a legal medical prescription. Drugs in this category are supervised due to their potential for misuse and substance use disorder. The classes of medications most commonly abused are opioids, central nervous system (CNS) depressants and central nervous stimulants. In particular, prescription opioid is most commonly abused in the form of prescription analgesics.

Harm reduction consists of a series of strategies aimed at reducing the negative impacts of drug use on users. It has been described as an alternative to the U.S.'s moral model and disease model of drug use and addiction. While the moral model treats drug use as a morally wrong action and the disease model treats it as a biological or genetic disease needing medical intervention, harm reduction takes a public health approach with a basis in pragmatism. Harm reduction provides an alternative to complete abstinence as a method for preventing and mitigating the negative consequences of drug use and addiction.

References

  1. 1 2 "FDA approves first buprenorphine implant for treatment of opioid dependence". U.S. Food and Drug Administration (FDA) (Press release). 26 May 2016. Retrieved 16 March 2017.
  2. "3 Patient Assessment". Clinical Guidelines for the Use of Buprenorphine in the Treatment of Opioid Addiction. Rockville (MD): Substance Abuse and Mental Health Services Administration (US). 2004.
  3. 1 2 "Commonly Used Terms". www.cdc.gov. 29 August 2017. Retrieved 16 July 2018.
  4. 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 American Psychiatric Association (2013), Diagnostic and Statistical Manual of Mental Disorders (5th ed.), Arlington: American Psychiatric Publishing, pp.  540–546, ISBN   978-0-89042-555-8
  5. 1 2 3 4 5 6 Substance Use and Mental Health Services Administration (30 September 2014). "Substance Use Disorders".
  6. 1 2 3 "Opioid Use and Opioid Use Disorder in Pregnancy". ACOG. August 2017. Retrieved 16 July 2018.
  7. 1 2 3 4 5 6 7 8 9 Sordo L, Barrio G, Bravo MJ, Indave BI, Degenhardt L, Wiessing L, Ferri M, Pastor-Barriuso R (April 2017). "Mortality risk during and after opioid substitution treatment: systematic review and meta-analysis of cohort studies". BMJ. 357: j1550. doi:10.1136/bmj.j1550. PMC   5421454 . PMID   28446428.
  8. 1 2 "Treatment for Substance Use Disorders". Substance Abuse and Mental Health Services Administration. October 2014.
  9. McDonald R, Strang J (July 2016). "Are take-home naloxone programmes effective? Systematic review utilizing application of the Bradford Hill criteria". Addiction. 111 (7): 1177–87. doi:10.1111/add.13326. PMC   5071734 . PMID   27028542.
  10. 1 2 Sharma B, Bruner A, Barnett G, Fishman M (July 2016). "Opioid Use Disorders". Child and Adolescent Psychiatric Clinics of North America. 25 (3): 473–87. doi:10.1016/j.chc.2016.03.002. PMC   4920977 . PMID   27338968.
  11. 1 2 3 4 5 Dydyk AM, Jain NK, Gupta M (2022), "Opioid Use Disorder", StatPearls, Treasure Island (FL): StatPearls Publishing, PMID   31985959, NCBI   NBK553166 , retrieved 16 November 2022
  12. Diagnostic and statistical manual of mental disorders : DSM-5. Arlington, VA : American Psychiatric Association. 2013. ISBN   978-0-89042-554-1.
  13. 1 2 3 4 Volkow ND, Koob GF, McLellan AT (January 2016). "Neurobiologic Advances from the Brain Disease Model of Addiction". The New England Journal of Medicine. 374 (4): 363–71. doi:10.1056/NEJMra1511480. PMC   6135257 . PMID   26816013. Addiction: A term used to indicate the most severe, chronic stage of substance-use disorder, in which there is a substantial loss of self-control, as indicated by compulsive drug taking despite the desire to stop taking the drug. In the DSM-5, the term addiction is synonymous with the classification of severe substance-use disorder.
  14. Webster LR (November 2017). "Risk Factors for Opioid-Use Disorder and Overdose". Anesthesia and Analgesia. 125 (5): 1741–1748. doi: 10.1213/ANE.0000000000002496 . PMID   29049118. S2CID   19635834.
  15. 1 2 3 4 5 Santoro TN, Santoro JD (2018). "Racial Bias in the US Opioid Epidemic: A Review of the History of Systemic Bias and Implications for Care". Cureus. 10 (12): e3733. doi: 10.7759/cureus.3733 . PMC   6384031 . PMID   30800543.
  16. 1 2 3 4 5 6 7 8 Dowell D, Ragan KR, Jones CM, Baldwin GT, Chou R (4 November 2022). "CDC Clinical Practice Guideline for Prescribing Opioids for Pain — United States, 2022". MMWR. Recommendations and Reports. 71 (3): 1–95. doi:10.15585/mmwr.rr7103a1. PMC   9639433 . PMID   36327391.
  17. "A Prescriber's Guide to Medicare Prescription Drug (Part D) Opioid Policies" (PDF).
  18. Mohamadi A, Chan JJ, Lian J, Wright CL, Marin AM, Rodriguez EK, von Keudell A, Nazarian A (August 2018). "Risk Factors and Pooled Rate of Prolonged Opioid Use Following Trauma or Surgery: A Systematic Review and Meta-(Regression) Analysis". The Journal of Bone and Joint Surgery. American Volume. 100 (15): 1332–1340. doi:10.2106/JBJS.17.01239. PMID   30063596. S2CID   51891341.
  19. "Prescription opioid use is a risk factor for heroin use". National Institute on Drug Abuse. October 2015. Retrieved 16 July 2018.
  20. 1 2 Hughes E (2 May 2018). "The Pain Hustlers". New York Times. Retrieved 3 May 2018.
  21. 1 2 "Trends in the Use of Methadone, Buprenorphine, and Extended-release Naltrexone at Substance Abuse Treatment Facilities: 2003–2015 (Update)". www.samhsa.gov. Retrieved 16 November 2022.
  22. Donovan DM, Ingalsbe MH, Benbow J, Daley DC (2013). "12-Step Interventions and Mutual Support Programs for Substance Use Disorders: An Overview". Social Work in Public Health. 28 (3–4): 313–332. doi:10.1080/19371918.2013.774663. PMC   3753023 . PMID   23731422.
  23. "Opioid Overdose Prevention Programs Providing Naloxone to Laypersons — United States, 2014". www.cdc.gov. Retrieved 16 November 2022.
  24. CDC (30 August 2022). "Disease of the Week – Opioid Use Disorder". Centers for Disease Control and Prevention. Retrieved 16 November 2022.
  25. "Data Brief 294. Drug Overdose Deaths in the United States, 1999–2016" (PDF). CDC. Retrieved 18 May 2018.
  26. Treatment Cf (2006). "[Table], Figure 4-4: Signs and Symptoms of Opioid Intoxication and Withdrawal". www.ncbi.nlm.nih.gov. Retrieved 6 April 2019.
  27. Fentanyl. Image 4 of 17. US DEA (Drug Enforcement Administration). See archive with caption: "photo illustration of 2 milligrams of fentanyl, a lethal dose in most people".
  28. 1 2 Kosten TR, Haile CN. Opioid-Related Disorders. In: Kasper D, Fauci A, Hauser S, Longo D, Jameson J, Loscalzo J. eds. Harrison's Principles of Internal Medicine, 19eNew York, NY: McGraw-Hill; 2014. http://accessmedicine.mhmedical.com/content.aspx?bookid=1130&sectionid=79757372 Accessed 9 March 2017.
  29. Diagnostic and statistical manual of mental disorders : DSM-5 (5th ed.). American Psychiatric Association. 2013. pp. 547–549. ISBN   978-0-89042-554-1.
  30. 1 2 Shah M, Huecker MR (2019). Opioid Withdrawal. StatPearls Publishing. PMID   30252268. NCBI   NBK526012 . Retrieved 21 October 2019.
  31. 1 2 3 4 5 6 7 8 9 10 11 Ries RK, Miller SC, Fiellin DA (2009). Principles of Addiction Medicine. Lippincott Williams & Wilkins. pp. 593–594. ISBN   978-0-7817-7477-2.
  32. Rahimi-Movaghar A, Gholami J, Amato L, Hoseinie L, Yousefi-Nooraie R, Amin-Esmaeili M (June 2018). "Pharmacological therapies for management of opium withdrawal". The Cochrane Database of Systematic Reviews. 2018 (6): CD007522. doi:10.1002/14651858.CD007522.pub2. PMC   6513031 . PMID   29929212.
  33. Chen KW, Banducci AN, Guller L, Macatee RJ, Lavelle A, Daughters SB, Lejuez CW (November 2011). "An examination of psychiatric comorbidities as a function of gender and substance type within an inpatient substance use treatment program". Drug and Alcohol Dependence. 118 (2–3): 92–9. doi:10.1016/j.drugalcdep.2011.03.003. PMC   3188332 . PMID   21514751.
  34. Webster LR, Webster RM (2005). "Predicting aberrant behaviors in opioid-treated patients: preliminary validation of the Opioid Risk Tool". Pain Medicine. 6 (6): 432–42. doi: 10.1111/j.1526-4637.2005.00072.x . PMID   16336480.
  35. Papaleontiou M, Henderson Jr CR, Turner BJ, Moore AA, Olkhovskaya Y, Amanfo L, Reid MC (July 2010). "Outcomes Associated with Opioid Use in the Treatment of Chronic Noncancer Pain in Older Adults: A Systematic Review and Meta-Analysis". Journal of the American Geriatrics Society. 58 (7): 1353–1369. doi:10.1111/j.1532-5415.2010.02920.x. PMC   3114446 . PMID   20533971.
  36. 1 2 Noble M, Tregear SJ, Treadwell JR, Schoelles K (February 2008). "Long-Term Opioid Therapy for Chronic Noncancer Pain: A Systematic Review and Meta-Analysis of Efficacy and Safety". Journal of Pain and Symptom Management. 35 (2): 214–228. doi: 10.1016/j.jpainsymman.2007.03.015 . PMID   18178367.
  37. Martell BA, O'Connor PG, Kerns RD, Becker WC, Morales KH, Kosten TR, Fiellin DA (16 January 2007). "Systematic Review: Opioid Treatment for Chronic Back Pain: Prevalence, Efficacy, and Association with Addiction". Annals of Internal Medicine. 146 (2): 116–127. doi:10.7326/0003-4819-146-2-200701160-00006. PMID   17227935. S2CID   28969290.
  38. Kalso E, Edwards JE, Moore AR, McQuay HJ (December 2004). "Opioids in chronic non-cancer pain: systematic review of efficacy and safety". Pain. 112 (3): 372–380. doi:10.1016/j.pain.2004.09.019. PMID   15561393. S2CID   25807828.
  39. 1 2 Goesling J, DeJonckheere M, Pierce J, Williams DA, Brummett CM, Hassett AL, Clauw DJ (May 2019). "Opioid cessation and chronic pain: perspectives of former opioid users". Pain. 160 (5): 1131–1145. doi:10.1097/j.pain.0000000000001493. PMC   8442035 . PMID   30889052.
  40. Krebs EE, Gravely A, Nugent S, Jensen AC, DeRonne B, Goldsmith ES, Kroenke K, Bair MJ, Noorbaloochi S (6 March 2018). "Effect of Opioid vs Nonopioid Medications on Pain-Related Function in Patients With Chronic Back Pain or Hip or Knee Osteoarthritis Pain: The SPACE Randomized Clinical Trial". JAMA. 319 (9): 872–882. doi: 10.1001/jama.2018.0899 . PMC   5885909 . PMID   29509867.
  41. Eriksen J, Sjøgren P, Bruera E, Ekholm O, Rasmussen NK (November 2006). "Critical issues on opioids in chronic non-cancer pain". Pain. 125 (1): 172–179. doi:10.1016/j.pain.2006.06.009. PMID   16842922. S2CID   24858908.
  42. Chaparro LE, Furlan AD, Deshpande A, Mailis-Gagnon A, Atlas S, Turk DC (April 2014). "Opioids Compared With Placebo or Other Treatments for Chronic Low Back Pain: An Update of the Cochrane Review". Spine. 39 (7): 556–563. doi:10.1097/BRS.0000000000000249. PMID   24480962. S2CID   25356400.
  43. Substitution maintenance therapy in the management of opioid dependence and HIV/AIDS prevention (PDF). World Health Organization. 2004. ISBN   978-92-4-159115-7.
  44. 1 2 "Treatment of opioid dependence". WHO. 2004. Archived from the original on 14 June 2010. Retrieved 28 August 2016.[ needs update ]
  45. Longo DL, Volkow ND, Koob GF, McLellan AT (28 January 2016). "Neurobiologic Advances from the Brain Disease Model of Addiction". New England Journal of Medicine. 374 (4): 363–371. doi:10.1056/NEJMra1511480. PMC   6135257 . PMID   26816013.
  46. Hyman SE (January 2007). "The neurobiology of addiction: implications for voluntary control of behavior". The American Journal of Bioethics. 7 (1): 8–11. doi:10.1080/15265160601063969. PMID   17366151. S2CID   347138.
  47. 1 2 3 4 5 6 7 Nestler EJ (December 2013). "Cellular basis of memory for addiction". Dialogues in Clinical Neuroscience. 15 (4): 431–43. doi:10.31887/DCNS.2013.15.4/enestler. PMC   3898681 . PMID   24459410. Despite the importance of numerous psychosocial factors, at its core, drug addiction involves a biological process.
  48. 1 2 3 Malenka RC, Nestler EJ, Hyman SE (2009). "Chapter 15: Reinforcement and Addictive Disorders". In Sydor A, Brown RY (eds.). Molecular Neuropharmacology: A Foundation for Clinical Neuroscience (2nd ed.). New York: McGraw-Hill Medical. pp. 364–375. ISBN   978-0-07-148127-4.
  49. 1 2 "Glossary of Terms". Mount Sinai School of Medicine. Department of Neuroscience. Archived from the original on 10 May 2019. Retrieved 9 February 2015.
  50. 1 2 Schultz W (July 2015). "Neuronal Reward and Decision Signals: From Theories to Data". Physiological Reviews. 95 (3): 853–951. doi:10.1152/physrev.00023.2014. PMC   4491543 . PMID   26109341.
  51. Brain & Behavior Research Foundation (13 March 2019). "The Biology of Addiction". YouTube.
  52. Robinson TE, Berridge KC (12 October 2008). "The incentive sensitization theory of addiction: some current issues". Philosophical Transactions of the Royal Society B: Biological Sciences. 363 (1507): 3137–3146. doi:10.1098/rstb.2008.0093. PMC   2607325 . PMID   18640920.
  53. 1 2 "Opioid Use Disorder".
  54. "Medically Supervised Withdrawal (Detoxification) from Opioids". pcssnow.org. 11 June 2021.
  55. Weiss RD (December 2011). "Adjunctive counseling during brief and extended buprenorphine-naloxone treatment for prescription opioid dependence: a 2-phase randomized controlled trial". Arch Gen Psychiatry. 68 (12): 1238–46. doi:10.1001/archgenpsychiatry.2011.121. PMC   3470422 . PMID   22065255.
  56. 1 2 Robison AJ, Nestler EJ (October 2011). "Transcriptional and epigenetic mechanisms of addiction". Nature Reviews. Neuroscience. 12 (11): 623–37. doi:10.1038/nrn3111. PMC   3272277 . PMID   21989194.
  57. 1 2 Olsen CM (December 2011). "Natural rewards, neuroplasticity, and non-drug addictions". Neuropharmacology. 61 (7): 1109–22. doi:10.1016/j.neuropharm.2011.03.010. PMC   3139704 . PMID   21459101.
  58. 1 2 Ruffle JK (November 2014). "Molecular neurobiology of addiction: what's all the (Δ)FosB about?". The American Journal of Drug and Alcohol Abuse. 40 (6): 428–37. doi:10.3109/00952990.2014.933840. PMID   25083822. S2CID   19157711.
  59. Blum K, Werner T, Carnes S, Carnes P, Bowirrat A, Giordano J, Oscar-Berman M, Gold M (2012). "Sex, drugs, and rock 'n' roll: hypothesizing common mesolimbic activation as a function of reward gene polymorphisms". Journal of Psychoactive Drugs. 44 (1): 38–55. doi:10.1080/02791072.2012.662112. PMC   4040958 . PMID   22641964.
  60. 1 2 Bourdy R, Barrot M (November 2012). "A new control center for dopaminergic systems: pulling the VTA by the tail". Trends in Neurosciences. 35 (11): 681–90. doi:10.1016/j.tins.2012.06.007. PMID   22824232. S2CID   43434322.
  61. 1 2 "Morphine addiction – Homo sapiens (human)". KEGG. Kanehisa Laboratories. 18 June 2013. Retrieved 11 September 2014.
  62. 1 2 Mistry C, Bawor M, Desai D, Marsh D, Samaan Z (May 2014). "Genetics of Opioid Dependence: A Review of the Genetic Contribution to Opioid Dependence". Current Psychiatry Reviews. 10 (2): 156–167. doi:10.2174/1573400510666140320000928. PMC   4155832 . PMID   25242908.
  63. Goldstein RZ, Volkow ND (October 2011). "Dysfunction of the prefrontal cortex in addiction: neuroimaging findings and clinical implications". Nature Reviews. Neuroscience. 12 (11): 652–69. doi:10.1038/nrn3119. PMC   3462342 . PMID   22011681.
  64. Schoenbum G, Shaham Y (February 2008). "The role of orbitofrontal cortex in drug addiction: a review of preclinical studies". Biol Psychiatry. 63 (3): 256–262. doi:10.1016/j.biopsych.2007.06.003. PMC   2246020 . PMID   17719014.
  65. Ieong HF, Yuan Z (1 January 2017). "Resting-State Neuroimaging and Neuropsychological Findings in Opioid Use Disorder during Abstinence: A Review". Frontiers in Human Neuroscience. 11: 169. doi: 10.3389/fnhum.2017.00169 . PMC   5382168 . PMID   28428748.
  66. 1 2 3 4 Nestler EJ (August 2016). "Reflections on: "A general role for adaptations in G-Proteins and the cyclic AMP system in mediating the chronic actions of morphine and cocaine on neuronal function"". Brain Research. 1645: 71–4. doi:10.1016/j.brainres.2015.12.039. PMC   4927417 . PMID   26740398. Specifically, opiates in several CNS regions including NAc, and cocaine more selectively in NAc induce expression of certain adenylyl cyclase isoforms and PKA subunits via the transcription factor, CREB, and these transcriptional adaptations serve a homeostatic function to oppose drug action. In certain brain regions, such as locus coeruleus, these adaptations mediate aspects of physical opiate dependence and withdrawal, whereas in NAc they mediate reward tolerance and dependence that drives increased drug self-administration.
  67. 1 2 "Opioid Use Disorder: Diagnostic Criteria". Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition (PDF). American Psychiatric Association. pp. 1–9. Archived from the original (PDF) on 26 November 2017. Retrieved 27 March 2017.
  68. Vargas-Perez H, Ting-A Kee R, Walton CH, Hansen DM, Razavi R, Clarke L, Bufalino MR, Allison DW, Steffensen SC, van der Kooy D (June 2009). "Ventral tegmental area BDNF induces an opiate-dependent-like reward state in naive rats". Science. 324 (5935): 1732–4. Bibcode:2009Sci...324.1732V. doi:10.1126/science.1168501. PMC   2913611 . PMID   19478142.
  69. Laviolette SR, van der Kooy D (March 2001). "GABA(A) receptors in the ventral tegmental area control bidirectional reward signalling between dopaminergic and non-dopaminergic neural motivational systems". The European Journal of Neuroscience. 13 (5): 1009–15. doi:10.1046/j.1460-9568.2001.01458.x. PMID   11264674. S2CID   46694281.
  70. 1 2 3 Nutt D, King LA, Saulsbury W, Blakemore C (March 2007). "Development of a rational scale to assess the harm of drugs of potential misuse". The Lancet. 369 (9566): 1047–1053. doi:10.1016/S0140-6736(07)60464-4. PMID   17382831. S2CID   5903121.
  71. 1 2 3 Dick DM, Agrawal A (2008). "The Genetics of Alcohol and Other Drug Dependence". Alcohol Res Health. 31 (2): 111–118. PMC   3860452 . PMID   23584813.
  72. Hall FS, Drgonova J, Jain S, Uhl GR (December 2013). "Implications of genome wide association studies for addiction: are our a priori assumptions all wrong?". Pharmacology & Therapeutics. 140 (3): 267–79. doi:10.1016/j.pharmthera.2013.07.006. PMC   3797854 . PMID   23872493.
  73. Bruehl S, Apkarian AV, Ballantyne JC, Berger A, Borsook D, Chen WG, Farrar JT, Haythornthwaite JA, Horn SD, Iadarola MJ, Inturrisi CE, Lao L, Mackey S, Mao J, Sawczuk A, Uhl GR, Witter J, Woolf CJ, Zubieta JK, Lin Y (February 2013). "Personalized medicine and opioid analgesic prescribing for chronic pain: opportunities and challenges". The Journal of Pain. 14 (2): 103–13. doi:10.1016/j.jpain.2012.10.016. PMC   3564046 . PMID   23374939.
  74. 1 2 Khokhar JY, Ferguson CS, Zhu AZ, Tyndale RF (2010). "Pharmacogenetics of drug dependence: role of gene variations in susceptibility and treatment". Annual Review of Pharmacology and Toxicology. 50: 39–61. doi:10.1146/annurev.pharmtox.010909.105826. PMID   20055697. S2CID   2158248.
  75. 1 2 Solhaug V, Molden E (17 October 2017). "Individual variability in clinical effect and tolerability of opioid analgesics - Importance of drug interactions and pharmacogenetics". Scand J Pain. 17: 193–200. doi:10.1016/j.sjpain.2017.09.009. PMID   29054049.
  76. "CDC Guideline for Prescribing Opioids for Chronic Pain | Drug Overdose | CDC Injury Center". www.cdc.gov. 31 August 2018. Retrieved 3 November 2018.
  77. "Our Commitment to Fight Opioid Abuse | CVS Health". CVS Health. Retrieved 3 November 2018.
  78. "Combat opioid abuse | Walgreens". Walgreens. Retrieved 3 November 2018.
  79. 1 2 "Naloxone for Treatment of Opioid Overdose" (PDF). U.S. Food and Drug Administration (FDA). Retrieved 7 November 2017.
  80. "Prevent Opioid Use Disorder | Drug Overdose | CDC Injury Center". www.cdc.gov. 31 August 2018. Retrieved 20 November 2018.
  81. "Naloxone". Human Metabolome Database – Version 4.0. 23 October 2017. Retrieved 2 November 2017.
  82. "Opioid Overdose Prevention Programs Providing Naloxone to Laypersons — United States, 2014". www.cdc.gov. Retrieved 9 March 2017.
  83. Childs R (July 2015). "Law enforcement and naloxone utilization in the United States" (PDF). U.S. Food and Drug Administration (FDA). North Carolina Harm Reduction Coalition. pp. 1–24. Retrieved 2 November 2017.
  84. "Case studies: Standing orders". NaloxoneInfo.org. Open Society Foundations. Archived from the original on 7 November 2017. Retrieved 2 November 2017.
  85. Schuckit MA (July 2016). "Treatment of Opioid-Use Disorders". The New England Journal of Medicine. 375 (4): 357–68. doi: 10.1056/NEJMra1604339 . PMID   27464203.
  86. Christie C, Baker C, Cooper R, Kennedy CP, Madras B, Bondi FA. The president's commission on combating drug addiction and the opioid crisis. Washington, DC, US Government Printing Office, November 2017;1.
  87. "Naloxone Opioid Overdose Reversal Medication". CVS Health. Retrieved 4 February 2019.
  88. 1 2 Morin KA, Vojtesek F, Acharya S, Dabous JR, Marsh DC (26 October 2021). "Evidence of Increased Age and Sex Standardized Death Rates Among Individuals Who Accessed Opioid Agonist Treatment Before the Era of Synthetic Opioids in Ontario, Canada". Cureus. 13 (10): e19051. doi: 10.7759/cureus.19051 . PMC   8608679 . PMID   34853762.
  89. Hser YI, Hoffman V, Grella CE, Anglin MD (May 2001). "A 33-Year Follow-up of Narcotics Addicts". Archives of General Psychiatry. 58 (5): 503–508. doi:10.1001/archpsyc.58.5.503. PMID   11343531.
  90. Horon IL, Singal P, Fowler DR, Sharfstein JM (June 2018). "Standard Death Certificates Versus Enhanced Surveillance to Identify Heroin Overdose–Related Deaths". American Journal of Public Health. 108 (6): 777–781. doi:10.2105/ajph.2018.304385. PMC   5944879 . PMID   29672148.
  91. Research Cf (2 March 2021). "Abuse-Deterrent Opioid Analgesics". FDA.
  92. Amato L, Davoli M, Minozzi S, Ferroni E, Ali R, Ferri M (28 February 2013). "Methadone at tapered doses for the management of opioid withdrawal". Cochrane Database of Systematic Reviews. 2013 (2): CD003409. doi:10.1002/14651858.CD003409.pub4. PMC   7017622 . PMID   23450540.
  93. MacArthur GJ, van Velzen E, Palmateer N, Kimber J, Pharris A, Hope V, Taylor A, Roy K, Aspinall E, Goldberg D, Rhodes T, Hedrich D, Salminen M, Hickman M, Hutchinson SJ (January 2014). "Interventions to prevent HIV and Hepatitis C in people who inject drugs: A review of reviews to assess evidence of effectiveness". International Journal of Drug Policy. 25 (1): 34–52. doi:10.1016/j.drugpo.2013.07.001. PMID   23973009.
  94. Nielsen S, Tse WC, Larance B (5 September 2022). Cochrane Drugs and Alcohol Group (ed.). "Opioid agonist treatment for people who are dependent on pharmaceutical opioids". Cochrane Database of Systematic Reviews. 2022 (9): CD011117. doi:10.1002/14651858.CD011117.pub3. PMC   9443668 . PMID   36063082.
  95. "Opioid substitution therapy or treatment (OST)". Migration and Home Affairs. European Commission. 14 March 2017.
  96. 1 2 3 4 5 6 7 8 9 Richard P. Mattick et al.: National Evaluation of Pharmacotherapies for Opioid Dependence (NEPOD): Report of Results and Recommendation
  97. Whelan PJ, Remski K (2012). "Buprenorphine vs methadone treatment: A review of evidence in both developed and developing worlds". Journal of Neurosciences in Rural Practice. 3 (1): 45–50. doi: 10.4103/0976-3147.91934 . PMC   3271614 . PMID   22346191.
  98. Martin JA, Campbell A, Killip T, Kotz M, Krantz MJ, Kreek MJ, McCarroll BA, Mehta D, Payte JT, Stimmel B, Taylor T, Wilford BB (October 2011). "QT Interval Screening in Methadone Maintenance Treatment: Report of aSAMHSA Expert Panel". Journal of Addictive Diseases. 30 (4): 283–306. doi:10.1080/10550887.2011.610710. PMC   4078896 . PMID   22026519.
  99. https://www.bccsu.ca/wp-content/uploads/2023/11/BC-OUD-Treatment-Guideline_2023-Update.pdf
  100. "MAT Medications, Counseling, and Related Conditions". www.samhsa.gov. Retrieved 10 November 2022.
  101. "Methadone". www.samhsa.gov. Retrieved 10 November 2022.
  102. 1 2 3 "Waiver Elimination (MAT Act)".
  103. 1 2 "Medication-Assisted Treatment (MAT)". www.samhsa.gov. Retrieved 9 November 2022.
  104. Gowing L, Farrell M, Ali R, White JM (May 2016). "Alpha₂-adrenergic agonists for the management of opioid withdrawal". The Cochrane Database of Systematic Reviews. 2016 (5): CD002024. doi:10.1002/14651858.CD002024.pub5. PMC   7081129 . PMID   27140827.
  105. Michels II, Stöver H, Gerlach R (2 February 2007). "Substitution treatment for opioid addicts in Germany". Harm Reduction Journal. 4 (1): 5. doi: 10.1186/1477-7517-4-5 . PMC   1797169 . PMID   17270059.
  106. "Many hospital policies create barriers to good management of opioid withdrawal". NIHR Evidence. 16 November 2022. doi:10.3310/nihrevidence_54639. S2CID   253608569.
  107. Harris M, Holland A, Lewer D, Brown M, Eastwood N, Sutton G, Sansom B, Cruickshank G, Bradbury M, Guest I, Scott J (14 April 2022). "Barriers to management of opioid withdrawal in hospitals in England: a document analysis of hospital policies on the management of substance dependence". BMC Medicine. 20 (1): 151. doi: 10.1186/s12916-022-02351-y . PMC   9007696 . PMID   35418095.
  108. Bonhomme J, Shim RS, Gooden R, Tyus D, Rust G (July 2012). "Opioid addiction and abuse in primary care practice: a comparison of methadone and buprenorphine as treatment options". Journal of the National Medical Association. 104 (7–8): 342–50. doi:10.1016/S0027-9684(15)30175-9. PMC   4039205 . PMID   23092049.
  109. Orman JS, Keating GM (2009). "Buprenorphine/naloxone: a review of its use in the treatment of opioid dependence". Drugs. 69 (5): 577–607. doi:10.2165/00003495-200969050-00006. PMID   19368419. S2CID   209147406.
  110. "Buprenorphine". www.samhsa.gov. Retrieved 10 November 2022.
  111. Yokell MA, Zaller ND, Green TC, Rich JD (1 March 2011). "Buprenorphine and Buprenorphine/Naloxone Diversion, Misuse, and Illicit Use: An International Review". Current Drug Abuse Reviews. 4 (1): 28–41. doi:10.2174/1874473711104010028. PMC   3154701 . PMID   21466501.
  112. "Medications for Opioid Use Disorder – Treatment Improvement Protocol 63". Substance Abuse and Mental Health Services Administration. Retrieved 20 November 2018.[ permanent dead link ]
  113. "What is Buprenorphine? | UAMS Psychiatric Research Institute". psychiatry.uams.edu/. Retrieved 17 November 2022.
  114. 1 2 3 4 Koehl JL, Zimmerman DE, Bridgeman PJ (18 July 2019). "Medications for management of opioid use disorder". American Journal of Health-System Pharmacy. 76 (15): 1097–1103. doi:10.1093/ajhp/zxz105. PMID   31361869.
  115. "Subutex and Suboxone Approved to Treat Opiate Dependence". U.S. Food and Drug Administration (FDA). 8 October 2002. Retrieved 1 November 2014.
  116. "Sublocade (buprenorphine) FDA Approval History". Drugs.com. Retrieved 10 November 2022.
  117. "SAMHSA Buprenorphine Quick Start Guide" (PDF).
  118. Wong JS, Nikoo M, Westenberg JN, Suen JG, Wong JY, Krausz RM, Schütz CG, Vogel M, Sidhu JA, Moe J, Arishenkoff S, Griesdale D, Mathew N, Azar P (December 2021). "Comparing rapid micro-induction and standard induction of buprenorphine/naloxone for treatment of opioid use disorder: protocol for an open-label, parallel-group, superiority, randomized controlled trial". Addiction Science & Clinical Practice. 16 (1): 11. doi: 10.1186/s13722-021-00220-2 . PMC   7881636 . PMID   33579359.
  119. "UpToDate".
  120. "About Opioid Addiction Treatment".
  121. "West Virginia Department of Human Health & Resources_Burea for Medical Services_Methadone" (PDF).
  122. "Methadone Side Effects: Common, Severe, Long Term". Drugs.com. Retrieved 10 November 2022.
  123. 1 2 "Vivitrol Prescribing Information" (PDF). Alkermes Inc. July 2013. Archived from the original (PDF) on 10 February 2017. Retrieved 2 November 2017.
  124. Skolnick P (January 2018). "The Opioid Epidemic: Crisis and Solutions". Annual Review of Pharmacology and Toxicology. 58 (1): 143–159. doi: 10.1146/annurev-pharmtox-010617-052534 . PMID   28968188.
  125. Sullivan MA, Garawi F, Bisaga A, Comer SD, Carpenter K, Raby WN, Anen SJ, Brooks AC, Jiang H, Akerele E, Nunes EV (December 2007). "Management of relapse in naltrexone maintenance for heroin dependence". Drug and Alcohol Dependence. 91 (2–3): 289–92. doi:10.1016/j.drugalcdep.2007.06.013. PMC   4153601 . PMID   17681716.
  126. Center for Substance Abuse Treatment (2009). Chapter 4—Oral Naltrexone. Substance Abuse and Mental Health Services Administration (US).
  127. Dalsbø TK, Steiro AK, Hammerstrøm KT, Smedslund G (2010). Heroin Maintenance for Persons with Chronic Heroin Dependence (Report). Oslo, Norway: Knowledge Centre for the Health Services at The Norwegian Institute of Public Health. PMID   29320074.
  128. Ferri M, Davoli M, Perucci CA (December 2011). "Heroin maintenance for chronic heroin-dependent individuals". The Cochrane Database of Systematic Reviews. 2011 (12): CD003410. doi:10.1002/14651858.CD003410.pub4. PMC   7017638 . PMID   22161378. S2CID   6772720.
  129. Rehm J, Gschwend P, Steffen T, Gutzwiller F, Dobler-Mikola A, Uchtenhagen A (October 2001). "Feasibility, safety, and efficacy of injectable heroin prescription for refractory opioid addicts: a follow-up study". Lancet. 358 (9291): 1417–23. doi:10.1016/S0140-6736(01)06529-1. PMID   11705488. S2CID   24542893.
  130. "Heroin Assisted Treatment | Drug Policy Alliance". Archived from the original on 16 March 2016. Retrieved 4 March 2016.
  131. Robertson JR, Raab GM, Bruce M, McKenzie JS, Storkey HR, Salter A (December 2006). "Addressing the efficacy of dihydrocodeine versus methadone as an alternative maintenance treatment for opiate dependence: A randomized controlled trial". Addiction. 101 (12): 1752–9. doi:10.1111/j.1360-0443.2006.01603.x. PMID   17156174.
  132. "Dihydrocodeine". Pubchem.
  133. "Login". online.lexi.com. Retrieved 2 November 2018.
  134. Carney T, Van Hout MC, Norman I, Dada S, Siegfried N, Parry CD (18 February 2020). "Dihydrocodeine for detoxification and maintenance treatment in individuals with opiate use disorders". The Cochrane Database of Systematic Reviews. 2 (2): CD012254. doi:10.1002/14651858.CD012254.pub2. PMC   7027221 . PMID   32068247.
  135. Ferri M, Minozzi S, Bo A, Amato L (June 2013). "Slow-release oral morphine as maintenance therapy for opioid dependence". The Cochrane Database of Systematic Reviews (6): CD009879. doi:10.1002/14651858.CD009879.pub2. PMID   23740540.
  136. "Bundesamt für Gesundheit – Substitutionsgestützte Behandlung mit Diacetylmorphin (Heroin)". Archived from the original on 13 March 2016.
  137. Tran TH, Griffin BL, Stone RH, Vest KM, Todd TJ (2017). "Methadone, Buprenorphine, and Naltrexone for the Treatment of Opioid Use Disorder in Pregnant Women". Pharmacotherapy: The Journal of Human Pharmaoclogy and Drug Therapy. 37 (7). Wiley: 824–839. doi:10.1002/phar.1958. PMID   28543191. S2CID   13772333.
  138. Zedler BK, Mann AL, Kim MM, Amick HR, Joyce AR, Murrelle EL, Jones HE (December 2016). "Buprenorphine compared with methadone to treat pregnant women with opioid use disorder: a systematic review and meta-analysis of safety in the mother, fetus and child". Addiction. 111 (12): 2115–2128. doi:10.1111/add.13462. PMC   5129590 . PMID   27223595.
  139. Kinsella M, Halliday LO, Shaw M, Capel Y, Nelson SM, Kearns RJ (29 July 2022). "Buprenorphine Compared with Methadone in Pregnancy: A Systematic Review and Meta-Analysis". Substance Use & Misuse. 57 (9): 1400–1416. doi: 10.1080/10826084.2022.2083174 . PMID   35758300.
  140. Suarez EA, Huybrechts KF, Straub L, Hernández-Díaz S, Jones HE, Connery HS, Davis JM, Gray KJ, Lester B, Terplan M, Mogun H, Bateman BT (1 December 2022). "Buprenorphine versus Methadone for Opioid Use Disorder in Pregnancy". New England Journal of Medicine. 387 (22): 2033–2044. doi:10.1056/NEJMoa2203318. PMC   9873239 . PMID   36449419.
  141. Suarez EA, Huybrechts KF, Straub L, Hernández-Díaz S, Jones HE, Connery HS, Davis JM, Gray KJ, Lester B, Terplan M, Mogun H, Bateman BT (December 2022). "Buprenorphine versus Methadone for Opioid Use Disorder in Pregnancy". New England Journal of Medicine. 387 (22): 2033–2044. doi:10.1056/NEJMoa2203318. PMC   9873239 . PMID   36449419.
  142. Meyer MC, Johnston AM, Crocker AM, Heil SH (2015). "Methadone and buprenorphine for opioid dependence during pregnancy: A retrospective cohort study". Journal of Addiction Medicine. 9 (2): 81–86. doi:10.1097/ADM.0000000000000092. PMC   4375023 . PMID   25622120.
  143. 1 2 3 4 5 6 Carley JA, Oesterle T (3 June 2021). "Therapeutic Approaches to Opioid Use Disorder: What is the Current Standard of Care?". Int J Gen Med. 14: 2305–2311. doi: 10.2147/IJGM.S295461 . PMC   8184146 . PMID   34113160.
  144. Dugosh K, Abraham A, Seymour B (March–April 2016). "A Systematic Review on the Use of Psychosocial Interventions in Conjunction With Medications for the Treatment of Opioid Addiction". J Addict Med. 10 (2): 93–103. doi:10.1097/ADM.0000000000000193. PMC   4795974 . PMID   26808307.
  145. Beck JS (18 August 2011). Cognitive behavior therapy : basics and beyond (Second ed.). New York: The Guilford Press. pp. 19–20. ISBN   978-1-60918-504-6. OCLC   698332858.
  146. Huibers MJ, Beurskens AJ, Bleijenberg G, van Schayck CP (July 2007). "Psychosocial interventions by general practitioners". The Cochrane Database of Systematic Reviews. 2007 (3): CD003494. doi:10.1002/14651858.CD003494.pub2. hdl: 2066/52984 . PMC   7003673 . PMID   17636726.
  147. 1 2 Longo DL, Schuckit MA (28 July 2016). "Treatment of Opioid-Use Disorders". New England Journal of Medicine. 375 (4): 357–368. doi: 10.1056/NEJMra1604339 . PMID   27464203.
  148. 1 2 3 4 Carroll KM (August 2005). "Behavioral therapies for drug abuse". Am J Psychiatry. 162 (8): 1452–60. doi:10.1176/appi.ajp.162.8.1452. PMC   3633201 . PMID   16055766.
  149. Vasilaki EI, Hosier SG, Cox WM (May 2006). "The Efficacy of Motivational Interviewing as a Brief Intervention for Excessive Drinking: A Meta-Analytic Review". Alcohol and Alcoholism. 41 (3): 328–335. doi: 10.1093/alcalc/agl016 . PMID   16547122.
  150. "Psychosocial interventions for opioid use disorder". UpToDate . Retrieved 2 November 2017.
  151. Dutra L (February 2008). "A meta-analytic review of psychosocial interventions for substance use disorders". Am J Psychiatry. 165 (2): 179–187. doi:10.1176/appi.ajp.2007.06111851. PMID   18198270. S2CID   29327821.
  152. Melemis SM (September 2015). "Relapse Prevention and the Five Rules of Recovery". The Yale Journal of Biology and Medicine. 88 (3): 325–32. PMC   4553654 . PMID   26339217.
  153. Sussman S (March 2010). "A review of Alcoholics Anonymous/ Narcotics Anonymous programs for teens". Evaluation & the Health Professions. 33 (1): 26–55. doi:10.1177/0163278709356186. PMC   4181564 . PMID   20164105.
  154. Galanter M (28 October 2016). "Combining medically assisted treatment and Twelve-Step programming: a perspective and review". The American Journal of Drug and Alcohol Abuse. 44 (2): 151–159. doi: 10.1080/00952990.2017.1306747 . PMID   28387530.
  155. Gamble J, O'Lawrence H (Summer 2016). "An Overview of the Efficacy of the 12-Step Group Therapy for Substance Abuse Treatment". Journal of Health and Human Services Administration. 39 (1): 142–160. PMID   27483978.
  156. 1 2 Donovan DM, Ingalsbe MH, Benbow J, Daley DC (26 August 2013). "12-Step Interventions and Mutual Support Programs for Substance Use Disorders: An Overview". Soc Work Public Health. 28 (3–4): 313–332. doi:10.1080/19371918.2013.774663. PMC   3753023 . PMID   23731422.
  157. Ward HB, Mosquera MJ, Suzuki J, Mariano TY (April 2020). "A Systematic Review of Noninvasive Brain Stimulation for Opioid Use Disorder". Neuromodulation. 23 (3): 301–311. doi:10.1111/ner.13108. PMID   32059080. S2CID   211122521.
  158. Young JR, Smani SA, Mischel NA, Kritzer MD, Appelbaum LG, Patkar AA (April–June 2020). "Non-invasive brain stimulation modalities for the treatment and prevention of opioid use disorder: a systematic review of the literature". J Addict Dis. 38 (2): 186–199. doi:10.1080/10550887.2020.1736756. PMID   32469286. S2CID   216247971.
  159. 1 2 Kock P, Froelich K, Walter M, Lang U, Dursteler KM (July 2022). "A systematic literature review of clinical trials and therapeutic applications of ibogaine". Journal of Substance Abuse Treatment. 138. doi:10.1016/j.jsat.2021.108717. PMID   35012793.
  160. Sullivan K (24 January 2023). "The psychedelic ibogaine can treat addiction. The race is on to cash in". The Guardian.
  161. "One Pill Can Kill". US Drug Enforcement Administration . Archived from the original on 15 November 2023. Retrieved 15 November 2023.
  162. "WHO | Information sheet on opioid overdose". WHO. Retrieved 6 April 2019.
  163. 1 2 GBD 2015 Mortality Causes of Death Collaborators (October 2016). "Global, regional, and national life expectancy, all-cause mortality, and cause-specific mortality for 249 causes of death, 1980–2015: a systematic analysis for the Global Burden of Disease Study 2015". Lancet. 388 (10053): 1459–1544. doi:10.1016/S0140-6736(16)31012-1. PMC   5388903 . PMID   27733281.
  164. 1 2 GBD 2013 Mortality Causes of Death Collaborators (January 2015). "Global, regional, and national age-sex specific all-cause and cause-specific mortality for 240 causes of death, 1990-2013: a systematic analysis for the Global Burden of Disease Study 2013". Lancet. 385 (9963): 117–71. doi:10.1016/S0140-6736(14)61682-2. PMC   4340604 . PMID   25530442.
  165. 1 2 "Understanding the Opioid Overdose Epidemic". cdc.gov. 8 August 2023.
  166. 1 2 3 Opioid Data Analysis and Resources. Drug Overdose. CDC Injury Center. Centers for Disease Control and Prevention. Click on "Rising Rates" tab for a graph. See data table below the graph.
  167. 1 2 American Society of Addiction Medicine. "Opioid Addiction 2016 Facts and Figures" (PDF).
  168. Health, United States, 2016: With Chartbook on Long-term Trends in Health (PDF). Hyattsville, MD.: CDC, National Center for Health Statistics. 2017. p. 4.
  169. Hempstead K (6 June 2013). "Supply-side response to declining heroin purity: fentanyl overdose episode in New Jersey". Health Economics. 23 (6): 688–705. doi:10.1002/hec.2937. PMID   23740651.
  170. 1 2 Ciccarone D (1 July 2022). "The Rise of Illicit Fentanyls, Stimulants and the Fourth Wave of the Opioid Overdose Crisis". Curr Opin Psychiatry. 34 (4): 344–350. doi:10.1097/YCO.0000000000000717. PMC   8154745 . PMID   33965972 via HHS Public Access.
  171. Manchikanti L, Singh VM, Staats PS, Trescot AM, Prunskis J (March 2022). "Fourth Wave of Opioid (Illicit Drug) Overdose Deaths and Diminishing Access to Prescription Opioids and Interventional Techniques: Cause and Effect". Pain Physician. 25 (2): 97–124. PMID   35322965.
  172. 1 2 Friedman J, Shover CL (December 2023). "Charting the fourth wave: Geographic, temporal, race/ethnicity and demographic trends in polysubstance fentanyl overdose deaths in the United States". Addiction. 118 (12): 2477–2485. doi: 10.1111/add.16318 . PMID   37705148.
  173. 1 2 Digital Communications Division (8 May 2018). "5-Point Strategy To Combat the Opioid Crisis". U.S. Department of Health and Human Services. Retrieved 2 November 2018.
  174. "Strategy to Combat Opioid, Abuse, Misuse, and Overdose" (PDF). U.S. Department of Health and Human Services. Retrieved 18 November 2018.
  175. 1 2 "Prescription Opioid Overdose Data". Center for Disease Control and Prevention. Retrieved 12 September 2016.
  176. 1 2 Paulozzi L (12 April 2012). "Populations at risk for opioid overdose" (PDF). U.S. Food and Drug Administration (FDA). Division of Unintentional Injury Prevention, National Center for Injury Prevention and Control Centers for Disease Control and Prevention. Retrieved 12 September 2016.
  177. "Opioid Addiction: 2016 Facts and Figures" (PDF). American Society of Addiction Medicine. Retrieved 12 September 2016.
  178. 1 2 "How Bad is the Opioid Epidemic?". PBS . Retrieved 12 September 2016.
  179. 1 2 Han B, Polydorou S, Ferris R, Blaum CS, Ross S, McNeely J (10 November 2015). "Demographic Trends of Adults in New York City Opioid Treatment Programs—An Aging Population". Substance Use & Misuse. 50 (13): 1660–7. doi:10.3109/10826084.2015.1027929. PMID   26584180. S2CID   5520930.
  180. "Facts & Faces of Opioid Addiction: New Insights". MAP Health Management. 2015. Archived from the original on 29 February 2020. Retrieved 12 September 2016.
  181. "Opioids". Substance Abuse and Mental Health Services Administration. 23 February 2016. Retrieved 12 September 2016.
  182. DeVencentis P (24 July 2017). "Grandson sold refrigerator for drugs, grandma says". USA Today.
  183. Peterman NJ, Palsgaard P, Vashi A, Vashi T, Kaptur BD, Yeo E, Mccauley W (2022). "Demographic and Geospatial Analysis of Buprenorphine and Methadone Prescription Rates". Cureus. 14 (5): e25477. doi: 10.7759/cureus.25477 . PMC   9246456 . PMID   35800815.
  184. "Use of Telemedicine While Providing Medication Assisted Treatment (MAT)" (PDF). SAMHSA. 15 May 2018. Retrieved 15 May 2018.
  185. Nunes EV, Levin FR, Reilly MP, El-Bassel N (March 2021). "Medication treatment for opioid use disorder in the age of COVID-19: Can new regulations modify the opioid cascade?". Journal of Substance Abuse Treatment. 122: 108196. doi:10.1016/j.jsat.2020.108196. PMC   7666540 . PMID   33221125.
  186. "Hochul signs legislation package to combat opioid crisis".
  187. "NY Decriminalizes Syringe Possession as Part of Overdose Prevention Efforts". 7 October 2021.
  188. "Products – Vital Statistics Rapid Release – Provisional Drug Overdose Data". 4 November 2021.
  189. 1 2 Abuse NI (20 January 2022). "Overdose Death Rates". National Institute on Drug Abuse. Retrieved 10 November 2022.
  190. 1 2 3 4 Overdose Death Rates. By National Institute on Drug Abuse (NIDA).
  191. Skolnick P (May 2022). "Treatment of overdose in the synthetic opioid era". Pharmacol Ther. 233. doi: 10.1016/j.pharmthera.2021.108019 . PMID   34637841.
  192. 1 2 Gomes T (2023). "Trends in Opioid Toxicity–Related Deaths in the US Before and After the Start of the COVID-19 Pandemic, 2011–2021". JAMA Netw Open. 6 (7): e2322303. doi:10.1001/jamanetworkopen.2023.22303. PMC   10329206 . PMID   37418260.
  193. "A qualitative assessment of circumstances surrounding drug overdose deaths during early stages of the COVID-19 pandemic". cdc.gov. August 2022.
  194. Simha S, Ahmed Y, Brummett CM (January 2023). "Impact of the COVID-19 pandemic on opioid overdose and other adverse events in the USA and Canada: a systematic review". Reg Anesth Pain Med. 48 (1): 37–43. doi:10.1136/rapm-2022-103591. PMID   36202619. S2CID   252753865.
  195. Jones CM, Shoff C (2023). "Fatal Drug Overdoses Among Medicare Beneficiaries Before and During the COVID-19 Pandemic". JAMA Psychiatry. 80 (5): 508–514. doi:10.1001/jamapsychiatry.2023.0310. PMC   10061313 . PMID   36988913.
  196. Moran KM (29 July 2022). "Changes in Racial/Ethnic Disparities in Opioid-Related Outcomes in Urban Areas during the COVID-19 Pandemic: A Rapid Review of the Literature". Int J Environ Res Public Health. 19 (15): 9283. doi: 10.3390/ijerph19159283 . PMC   9368442 . PMID   35954640.
  197. "Prescribing controlled substances via telehealth". 16 October 2023.
  198. James W. Kalat, Biological Psychology. Cengage Learning. Page 81.
  199. EMEA 19 April 2001 EMEA Public Statement on the Recommendation to Suspend the Marketing Authorisation for Orlaam (Levacetylmethadol) in the European Union Archived 4 February 2017 at the Wayback Machine
  200. US FDA Safety Alerts: Orlaam (levomethadyl acetate hydrochloride) Page Last Updated: 20 August 2013 Archived 18 January 2017 at the Wayback Machine

Further reading