Abstract
The opioid epidemic is at the epicenter of the drug crisis with an unimaginable number of overdose deaths and exorbitant associated medical costs that have crippled many communities throughout the socioeconomic spectrum in the US. Classic medications for the treatment of opioid use disorders predominantly target the opioid system and thus unfortunately have been underutilized in part due to their own abuse potential and heavy regulatory burden for patients and clinicians. Opioid antagonists are now evolving in use not only to prevent acute overdoses but as extended use treatment options. Strategies that target specific genetic and epigenetic factors and novel non-opioid medications hold promise as future therapeutic interventions of opioid abuse. Success in increasing the treatment options in the clinical toolbox will hopefully help break the historical pattern of recurring opioid epidemics.
Introduction
The scourge of drug abuse has often defined key periods of human history. 175 years ago, when the first issue of what would become the American Journal of Psychiatry appeared, the Opium Wars dominated life in Asia (1, 2). Opium, which initially been imported into China for medicinal purposes, had quickly transitioned to recreational use and addiction that penetrated into much of the region devastating all levels of society. As the Chinese emperors attempted to halt the epidemic, Western nations fought the Chinese to increase opium imports and taxes Another heroin epidemic, this time particularly affecting urban cities in the United States in the 1970s and American veterans of the Vietnam war served as the major impetus for the creation of the Drug Enforcement Administration (DEA) in 1973 (3). Today, the scourge of a new wave of opioid addiction has transcended every sociodemographic community in the U.S. leading to severe healthcare and societal burden of epidemic proportions with an economic cost of over $500 billion per year (4).
The opioid tsunami that has gripped the country stemmed in large part from a distorted and biased understanding of addiction vulnerability, fueled by a fervent over-prescription of opioid analgesics which yearly exceeded the clinical needs of the entire adult population in the USA. The broad exposure to potent opioids, across the socioeconomic spectrum, led many individuals to heroin (approximately 80% of new heroin users started out misusing opioid prescription analgesics)(5–7), or the illegal and cheaper version of prescription medications such as fentanyl whose popularity and illegal sales increased after federal regulations reduced access to legal prescriptions. The consequences have been shocking with over 50,000 overdose deaths yearly (8, 9), a number expected to continue in coming years if drastic interventions are not taken. The devastating impact of the opioid epidemic has had profound medical consequences with an approximate 3000% rise in medical services needed for patients with opioid misuse and dependence as evident with an increase of ~217,000 patients provided medical care in 2007 to approximately 7 million in 2014 (10).
Despite the significant need for therapeutic interventions to meet the urgent demands of the opioid crisis, most of the over 2.6 million people diagnosed with an opioid use disorder (OUD) receive minimal treatment for their addiction. The most common pharmacotherapies for OUDs are opioid substitution medications that paradoxically bear marked stigma and tight governmental regulations due to their abuse liability and potential diversion to the black market. Additionally, these medications require very close clinical monitoring that altogether incur a significant healthcare burden. Thus, together with the scarcity of clinicians trained in the recognition and treatment of substance use disorders, the ability of the current treatment system has been limited to service the enormous proportions of people needed to be treated in the current epidemic. We submit that a multi-pronged strategy including a broad repertoire of treatment options based on science-driven approaches are critical to meet not only the current epidemic, but also to prevent future outbreaks. We summarize below current opioid therapeutic strategies and explore some of the diverse and unique approaches being developed to expand the clinical toolbox for treating OUD.
Current Strategies of Opioid treatments
Addiction is a chronic brain disorder that requires long term treatment. Disturbingly, commercials touting an expensive addiction “cure” after 30 days in a spa-like residential program receiving group therapy reflect an abysmal lack of knowledge of the abundant clinical research literature. Such abstinence only residential treatment programs, despite the promise of a “cure,” have very high relapse rates shortly after “graduation” or discharge. Medication assisted treatment (MAT) has the best long term results and for opioid use disorder; there are currently several different medication options.
The full agonist approach is represented by methadone. This treatment was developed in the 1960s when it was discovered that opioid-addicted patients could be maintained on a single daily dose of methadone with reduction in craving and drug-seeking behavior. Over 50 years of data have demonstrated that correctly treated patients on methadone plus counseling are able to function well in school or employment and maintain a good quality life. Tolerance develops to all opioid agonists and methadone is no exception. But tolerance does not continue to increase so methadone can be prescribed at the same dose for many years. Problems, however, do arise when the medication is stopped since detoxification can be difficult and take many months.
The partial agonist approach is represented by buprenorphine. This medication has high affinity for the mu opioid receptor (MOR), but has an upper limit or “ceiling” on maximal opioid effects. It blocks craving and drug-seeking similar to methadone, but its limited ceiling effect means that patients with a very high pharmacological level of opioid dependence may not be able to be transferred directly to buprenorphine. In the United States, a combination medication is generally used consisting of buprenorphine and Naloxone (Suboxone). If the combination is injected, instead of ingested by the normal oral/sublingual route, naloxone reduces the pleasurable effects of MOR stimulation and thus discourages abuse.
A recent treatment innovation is the development of several extended release injectable formulations. One that provides slow release of buprenorphine for 30 days is expected to be marketed beginning in 2018. Others lasting as long as 6 months are currently under FDA review.
The antagonist approach is represented by naltrexone. The oral version was approved by the FDA in 1985. It occupies opioid receptors and prevents agonist drugs such as heroin or methadone from binding to the receptors and as a pure antagonist does not produce euphoria or reward. The oral version requires daily or three times weekly administration but patients can relapse simply by stopping the medication for 48 hours. Thus, the oral form of naltrexone had very limited success. More recently, an extended release version of naltrexone has become available. This version prevents relapse to opioid addiction for 30 days. Many patients find it convenient to return monthly for an injection rather than to take a daily medication. In a 2016 clinical trial in volunteer patients in the probation system, those randomly assigned to 6 months on extended release naltrexone had significantly more drug-negative urines and a lower relapse rate than patients given usual treatments in the community (11). Antagonist treatments are currently not as yet widely accepted. It is considered challenging to integrate them into the normal opioid agonist treatment regimens because detoxification of the patients is required before an antagonist can be administered. The fact that initial detoxification normally occurs in residential treatment does an important clinical window in which antagonist treatment can be initiated before individuals leave the protected environment.
Overall, looking back over the course of the opioid epidemic has highlighted several challenges with conventional opioid medications that need to be considered in trying to change the trajectory of this crisis. First, very few physicians were trained in the biology of addiction and the use of opioid medications. As such, existing opioid treatments are still not optimally used in treating pain. Second, there continues to be a bias toward opioid agonists for initial treatment. While this is an important option particularly for OUD individuals maintained for years on these agonists, newly afflicted individuals are rarely given the opportunity to be treated with the extended release antagonists that are effective and devoid of the addictive properties associated with opioid agonists. Moreover, limited non-opioid strategies exist.
LOOKING FORWARD TO DIFFERENT APPROACHES
A number of new therapeutic strategies are currently being explored that might help to expand current ways of thinking to eventually accelerate the development of effective interventions.
Genetic Strategies in opioid treatment – Pharmacogenomics
Individual differences in relation to genetics play an important role in OUD vulnerability. It is estimated based on twin studies that approximately 50% of the variation in opioid addiction is attributed to genetic factors (12, 13). While genetics is not deterministic for developing a substance use disorder, especially if the person is never exposed to the agent, knowledge regarding genetic vulnerability can help provide important insights regarding the underlying neurobiology of the disorder, reveal novel biological target for potential therapeutic development, and potentially optimize personalized medication therapy. The OPRM1 gene on chromosome 6 that encodes the mu opioid receptor (MOR) has logically been a high-priority candidate for studies investigating disease risk and pharmacogenomic factors of opioid use. The locus of the OPRM1 gene that has received most attention is the common missense single-nucleotide polymorphism (SNP) A118G rs1799971, a nonsynonymous point mutation that changes the amino acid sequence of the protein (14). The OPRM1 variants have been demonstrated to have functional relevance in relation to in vitro MOR binding and signaling (15–17), in vivo MOR binding (18, 19), MOR signaling in human postmortem specimens (20–22) and opioid neuropeptide gene expression levels in the human brain relevant to addiction (23). Most of the findings suggest reduced MOR in A118G subjects. Other OPRM1 variants have also been investigated in relation to heroin addiction (24–27) and the functional relationship to MOR signaling and downstream transcriptional regulation (21).
Multiple studies have addressed the relationship of the rs1799971 polymorphisms to heroin/opioid abuse (15, 23, 28–30). Not surprisingly the OPRM1 results from the candidate gene studies to date have been equivocal due in part to low sample sizes, differences in race and ethnicity or potential phenotype/environmental variables among other factors. Meta-analysis studies that attempt to increase the statistical power by combining the results from multiple investigations have also been inconclusive regarding OUD (31, 32) but suggest a contribution to addiction liability shared across different addictive substances (33). There is also research implicating the rs1799971 allele in naltrexone response in the treatment of alcohol use disorder (34, 35). Based particularly on the multifaceted nature of addiction it is evident that a single gene is an extremely limited strategy to demonstrate conclusive genetic contributions. Indeed, a large comprehensive replication study demonstrated that the rs1799971 SNP was only associated with heroin addiction in the presence of another SNP (rs3778150), which had been identified as a disease-associated expression quantitative trait loci (eQTL) that influenced OPRM1 expression in the human prefrontal cortex (26). These finding may explain some of the discrepant literature regarding the association between the rs1799971 genotype and heroin/opioid addiction and also highlights the importance of haplotype strategies for complex disorders like addiction, where the combination of alleles that are inherited together has stronger statistical power in associating a genetic link with the phenotype.
An important question for guiding future clinical care is whether documented functional differences of OPRM1 variants could be leveraged to improve the pharmacological response in patients undergoing opioid treatment (e.g., methadone) and to prevent adverse effects including addiction vulnerability in healthy individuals being prescribed opioid analgesics. Determining the effective individual dose for methadone is often clinically challenging since under-dosing can lead to craving and relapse and high doses can induce euphoria and sedation as well as other side effects. Implementing an agnostic genome-wide association study (GWAS) approach Gelernter and colleagues (36) recently identified one statistically significant region in the genome that was associated with higher daily methadone dosing in opioid-dependent African-Americans (but not European-Americans) patients. Interestingly, the region was on chromosome 6 with the lead SNP rs73568641 localized in the OPRM1 gene. The authors replicated the finding showing the SNP associated with increased morphine dose requirement for pain relief in an independent sample of African-American surgical patients. Significant research remains to be conducted to determine whether the rs73568641 SNP has a causal relationship to the expression or function of the MOR. Nevertheless, the findings are a critical step forward suggesting that OPRM1 genetics could be potentially useful clinically in determining appropriate opioid medication dose. Recent meta-analysis (37) and other studies (38, 39) also suggest that the A118G rs1799971 allele variant can influence opioid pain management with individuals carrying the A118G rs1799971 allele requiring higher opioid doses than A118A subjects. The fact that the OPRM1 might hold promise as a genetic predictor of opioid medication dose in the setting of addiction treatment and in analgesia could be potentially helpful in identifying non-dependent individuals who might be at potential addiction risk when being treated with opioid prescription medications. Large-scale investigations still, however, are needed before individual OPRM1 genetics can be incorporated into the clinical formula in the future for setting optimal opioid treatment dosage in OUD and pain management.
It is also important to reemphasize that it is unlikely that only the OPRM1 gene will be able to inform and improve clinically relevant treatment based on genetics. Functional genetic variations of other genes such as those involved with liver metabolic enzyme activity were recently reported to associate with the steady-state plasma concentration of methadone enantiomers, which provide a measure of methadone metabolism and are used clinically as an index of treatment response and efficacy of methadone therapy (40, 41). If replicated, such strategies will help individualize treatment to achieve dose optimization for OUD patients to reduce and avert the onset of withdrawal symptoms as well as to optimize opioid pain management for non-dependent subjects.
Alternative Splicing to Guide Targeted Opioid Medications
DNA sequence variations and the mechanism of their regulation of gene expression and disease phenotype are complex and not well understood, but multiple processes have begun to be explored as potential targets for medication development. Alternative splicing of genes is an efficient means of generating variation in protein function and thus has been of growing interest in attempts to personalize and optimize pharmacological therapies. Splicing determine which of a gene’s exons that code for its amino acid product, i.e., the mu opioid receptor, are used or not used to synthesize the final receptor. As a result, there can be multiple subtypes of the mu receptor, based on differences in splicing. Not surprising, the development of novel medications based on molecular genetics has also involved consideration of the multiple isoforms of the MOR. An array of MOR variants are produced by alternative pre-mRNA splicing of the single-copy of the OPRM1 gene (42, 43). The extensive alternative splicing of OPRM1 creates at least three structurally distinct classes of splice variants that are conserved from rodent to human thus improving the possibility for preclinical scientific studies to better inform human investigations. Animal studies have shown, for example, that the different truncated variants at the C-termini generated from 3′ alternative splicing of the OPRM1 gene do not substantially affect morphine analgesia, but differentially alter morphine-induced tolerance, physical dependence and reward behavior (44). Additionally, whereas normal analgesia is maintained for morphine and methadone analgesia in variants within exon 11 of the OPRM1 gene, the analgesic actions of heroin and fentanyl are markedly decreased (45). Thus, developing opioid analgesics that lack the side effects of traditional opioids may be possible by targeting truncated splice variants of the MOR (46, 47). Altogether, research efforts to dissociate the desirable analgesic properties of opioids from undesirable side effects of addiction appear possible. Targeting specific regions of the MOR could be an effective therapeutic strategy to reduce the abuse and addiction liability of opioids while maintaining analgesic properties.
The recent selective molecular targeting of the MOR through biased agonism, though not a genetic approach, is also a significant advance in being able to selectivity target specific downstream signal transduction pathways in the same G-protein coupled receptor (GPCR) for medication development (48–50). In contrast to the classic categorization of ligands as full, partial or inverse agonists or antagonists, biased agonism leverages the capability of GPCRs to stabilize receptor conformation to regulate different signaling pathways. As such agonists have been designed to deliver different physiologic outcomes by biasing a selective downstream signal transduction pathway (such as G-protein signaling, beta-arrestin recruitment and receptor internalization) mediated by the same receptor. This strategy significantly expands the repertoire for drug discovery for ligands targeting MOR signaling to potentially have analgesic properties (such as those recruiting beta-arrestin proteins) while avoiding tolerance or other opioid adverse effects (linked to G-protein signaling) (51, 52). Clearly, the fact that individual variation exist for genes aligned to distinct GPCR pathways indicates that genetic factors might also dictate which individuals might respond to certain biased agonists.
Epigenetics inform opioid treatment
In addition to genetics, susceptibility to opioid addiction is known to be strongly influenced by environmental factors. As such epigenetics—biological mechanisms that mediate genetic control of gene expression without a change in DNA sequence—could be of significant importance for understanding individual vulnerability to addiction and response to treatment. The epigenetic mechanisms that turn on and off genes to set the state of gene expression patterns and thus cellular function include methylation of DNA and modifications (e.g., methylation, acetylation, phosphorylation) of histones, around which DNA is bound that together constitutes chromatin. Epigenetics has emerged as an important biological driver of addiction pathology (53–56). Most epigenetic studies to date relevant to OUD have focused on DNA methylation. A number of investigations have reproducibly observed that chronic exposure to opioids (chronic opioid-treated pain patients, active heroin abusers or former heroin users undergoing methadone maintenance) induce epigenetic changes in peripheral marks (lymphocyte and blood) including increased methylation of the OPRM1 gene (57–59) (60, 61). The hypermethylation of DNA located in the OPRM1 promoter appears to block the binding of transcription activators such as Sp1 which ultimately leads to silencing of the OPRM1 (62). Reduced MOR expression that has been detected in various brain regions of heroin abusers (21, 63, 64) might relate to their increased opioid requirement. Consistently, pain relief in cancer patients has been shown to correlate with methylation of the OPRM1 promoter with high-dose opioid use associated with OPRM1 hypermethylation (57). These and other studies suggest that DNA methylation in peripheral blood samples, and thus a potential proxy for CNS MOR function, could provide a biomarker for OPRM1 function that could aide in determining opioid dosage. It is, however, important to emphasize the cell-specific nature of epigenetic mechanisms where clear DNA methylation differences have recently been revealed in different neurons and glia in the prefrontal cortex of heroin abusers (65), so it is unclear what specific CNS function alterations of peripheral OPRM1 methylation would predict. Additionally, while the OPRM1 is a rationale target for research in guiding future clinical care, the gene list needs to be expanded based on gathering genome-wide unbiased data from large scaled clinical studies to more efficiently direct pharmacoepigenetic approaches.
A critical aspect of epigenetics that makes it an intriguing strategic therapeutic target is that the modifications are reversible. Moreover, multiple families of proteins are involved in adding (writers), recognizing (readers) or removing (erasers) epigenetic marks (66, 67). This plethora of proteins provides a diverse system to tweak the tone of gene expression and thus cellular functions and phenotypes relevant to addiction. The importance of epigenetic to OUD was highlighted in a recent postmortem interrogation of the striatum of human heroin abusers (53). Epigenetic disturbances were observed to correlate with alterations of genes relevant to glutamatergic function and synaptic plasticity, impairments of which are well acknowledged as a hallmark of addiction pathology (68, 69). Interestingly, enhanced histone acetylation levels (and specifically acetylation of histone H3 protein, lysine 27) in the striatum of abusers correlated significantly with the years of heroin use. It is well known that acetylated-lysine residues on chromatin are specifically recognized and ‘read’ by the BET (bromodomains and extra-terminal) subfamily of proteins. BET inhibitors have become a popular strategy developed as anti-cancer medications that could provide novel agents to repurpose as potential treatments for OUD. A small molecular BET inhibitor, JQ-1, reduced heroin self-administration and heroin-seeking behavior in a rodent model thus setting the stage for BET inhibitors to be investigated in clinical trials in subjects with OUD. The wide range of epigenetic molecules being developed for many clinical symptoms and diseases opens a treasure trove of compounds that could be examined in relation to epigenetic pathlogies in addiction.
Medical Cannabinoid — Cannabidiol
Recent attention has focused on so-called “medical marijuana” as a potential non-conventional strategy. While still in their infancy in gathering data, some epidemiological studies have recently emerged suggesting that in states with medical marijuana laws there has been a reduction in opioid-related deaths, opioid prescriptions and opioid-related car fatalities (70–74). Many reasons, even those unrelated to the pharmacology of cannabis on brain function relevant to opioid use, might account for the apparent associations. It is, however, clear that the broad usage of the term “medical marijuana” (often confused with conventional recreational marijuana) ignores the complex nature of the plant with hundreds of cannabinoids and other entourage chemicals essential to consider in the development of a clinically useful medication. What is known from a number of preclinical studies is that different cannabinoids can have adverse or beneficial effects on opioid sensitivity. For example, whereas THC, the psychoactive component of cannabis, can enhance the reward sensitivity to opioids (75–78), the exposure to cannabidiol (CBD), a non-rewarding cannabinoid, reduces the reward-facilitating effect of morphine (79) and reduces cue-induced heroin-seeking behavior even weeks following the last CBD exposure (80). CBD normalizes glutamatergic impairments induced by heroin self-administration (80). Such findings have set in motion many research studies evaluating not only opioids, but other drugs of abuse in relation to the potential impact of CBD. Moreover, results from pilot clinical studies have suggested replication of the animal findings with CBD reducing cue-induced craving, as well as anxiety, in heroin-abstinent subjects (81). Intriguingly, similar to the rodent model, CBD maintained a reduction of heroin craving even a week after its last administration. The protracted effects of CBD might be of particular benefit for a successful therapeutic strategy for OUD since it could maintain protective effects to reduce craving and thus relapse even if the individual missed a daily dose. Importantly, CBD lacks any rewarding effects (79, 82–85), has a wide safety margin (86–88) and thus would not require the restrictive governmental regulations associated with opioid agonist medications that have abuse potential and are diverted to the black market. However, CBD is still currently under the cannabis umbrella of a Schedule I drug. As additional clinical trials are conducted, the knowledge gained will hopefully help revise the federal regulations so that a full battery of research can be explored to determine the potential of CBD for OUD treatment. Similar to all other novel strategies, the future application of “medicinal Cannabidiol” for OUDs needs to determine what specific aspect of the complex clinical spectrum of the disorder (e.g., craving versus acute reward substitution) this approach would be most optimal to target.
Vaccines
Although not new, another “outside the box” approach that originally had been considered nearly 40 years ago involves the development of anti-drug vaccines. The first vaccine was designed to target opioids (89). Vaccines for the treatment of other drugs of abuse such as nicotine (90–92) and cocaine (93–96) have been tested to a greater extent in human subjects with mixed results seeming to depend on individual variability in antibody titer levels. The challenge of raising sufficiently high antibody titers has been recently addressed with a novel strategy to develop a more efficacious heroin conjugate vaccine in combination with specific carrier proteins and adjuvants (97, 98). This anti-heroin vaccine approach was recently evaluated in pre-clinical models (mice and non-human primates) and resulted in a significant 15-fold reduction of heroin operant responding for 8 months in non-human primates (99). Future clinical studies will help to determine whether the promise of anti-heroin vaccines can indeed achieve their long promise.
CONCLUSION: Steps to move forward/roadmap forward
We cannot address the current opioid epidemic with old tools including declarations of an opioid ‘war’ and harsh judicial ramifications as previously employed over the past century. They failed in the past and exacerbated psychosocial pathologies that persists today. Instead, it is essential that education of prescribing physicians and the general public about the benefits and dangers of opioids are complemented with the rapid development and translation of novel strategies to expand the currently available medications. To meet an epidemic, a different mentality needs to be employed where specific paths are created at the level of the federal and state governments to mobilize the efforts of scientists and clinicians to advance care, prevention and ultimately treatments. Strategies should span the improvement of current opioid treatments by leveraging genetic and epigenetic factors as well as the development of new therapies such as medical cannabinoids and innovative medications that could specifically strengthen impaired synaptic plasticity in the management of OUD. These approaches might also be employed to reduce the transition to addiction in non-dependent patients administered prescription opioids for chronic pain.
What continues to be missing in the development of novel medications, especially in consideration of personalized medicine and the complex nature of addiction disorders, is structured phenotyping of patients on which to integrate genetic and epigenetic data. Such knowledge can provide a strong biological foundation on which to truly develop better targeted personalized medication strategies. Nevertheless, irrespective of developing the most effective innovative medication for OUD, supportive social services must go hand in hand with drug development. There will not be a miracle therapeutic strategy. The science-based future medication approaches discussed above and in other publications are interesting, but even the most promising will fail to be realized without fast-track transition of preclinical and early stage phase I clinical studies to full clinical trials and incorporating an “all hands on board” approach that even involves input from patients and families. There is much to be learned after 175 years on which to transform the medication clinical toolkit in coming years.
References
- 1.Lovell J. The Opium War: Drugs, Dreams, and the Making of Modern China. The Overlook Press: 2015. [Google Scholar]
- 2.Fay P. The Opium War, 1840-1842: barbarians in the Celestial Empire in the early part of the nineteenth century and the war by which they forced her gates ajar. 2000 [Google Scholar]
- 3.The DEA Years: 1970-1975. https://www.dea.gov/about/history.shtml.
- 4.Advisers TCoE: The Underestimated Cost of the Opioid Crisis. 2017 [Google Scholar]
- 5.Muhuri P, Gfroerer J, Davies C. Associations of nonmedical pain reliever use and initiation of heroin use in the United States. CBHSQ Data Review. http://archive.samhsa.gov/data/2k13/DataReview/DR006/nonmedical-pain-reliever-use-2013.pdf2013.
- 6.Banerjee G, Edelman EJ, Barry DT, Becker WC, Cerda M, Crystal S, Gaither JR, Gordon AJ, Gordon KS, Kerns RD, Martins SS, Fiellin DA, Marshall BD. Non-medical use of prescription opioids is associated with heroin initiation among US veterans: a prospective cohort study. Addiction. 2016;111:2021–2031. doi: 10.1111/add.13491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Compton WM, Jones CM, Baldwin GT. Relationship between Nonmedical Prescription-Opioid Use and Heroin Use. N Engl J Med. 2016;374:154–163. doi: 10.1056/NEJMra1508490. [DOI] [PubMed] [Google Scholar]
- 8.Provisional Counts Of Drug Overdose Deaths, as of 8/6/2017. CDC •National Center for Health Statistics• National Vital Statistics System. 2017 [Google Scholar]
- 9.Ahmad F, Rossen L, Spencer M, Warner M, Sutton P. Provisional drug overdose death counts CDC/National Center for Health Statistics. 2018 [Google Scholar]
- 10.The Impact of the Opioid Crisis on the Healthcare System. A FAIR Health White Paper. FAIR Health; 2016. [Google Scholar]
- 11.McDonald RD, Tofighi B, Laska E, Goldfeld K, Bonilla W, Flannery M, Santana-Correa N, Johnson CW, Leibowitz N, Rotrosen J, Gourevitch MN, Lee JD. Extended-release naltrexone opioid treatment at jail reentry (XOR) Contemp Clin Trials. 2016;49:57–64. doi: 10.1016/j.cct.2016.05.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Tsuang MT, Lyons MJ, Meyer JM, Doyle T, Eisen SA, Goldberg J, True W, Lin N, Toomey R, Eaves L. Co-occurrence of abuse of different drugs in men: the role of drug-specific and shared vulnerabilities. Arch Gen Psychiatry. 1998;55:967–972. doi: 10.1001/archpsyc.55.11.967. [DOI] [PubMed] [Google Scholar]
- 13.Kendler KS, Jacobson KC, Prescott CA, Neale MC. Specificity of genetic and environmental risk factors for use and abuse/dependence of cannabis, cocaine, hallucinogens, sedatives, stimulants, and opiates in male twins. Am J Psychiatry. 2003;160:687–695. doi: 10.1176/appi.ajp.160.4.687. [DOI] [PubMed] [Google Scholar]
- 14.Huang P, Chen C, Mague SD, Blendy JA, Liu-Chen LY. A common single nucleotide polymorphism A118G of the mu opioid receptor alters its N-glycosylation and protein stability. Biochem J. 2012;441:379–386. doi: 10.1042/BJ20111050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Bond C, LaForge KS, Tian M, Melia D, Zhang S, Borg L, Gong J, Schluger J, Strong JA, Leal SM, Tischfield JA, Kreek MJ, Yu L. Single-nucleotide polymorphism in the human mu opioid receptor gene alters beta-endorphin binding and activity: possible implications for opiate addiction. Proc Natl Acad Sci U S A. 1998;95:9608–9613. doi: 10.1073/pnas.95.16.9608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Deb I, Chakraborty J, Gangopadhyay PK, Choudhury SR, Das S. Single-nucleotide polymorphism (A118G) in exon 1 of OPRM1 gene causes alteration in downstream signaling by mu-opioid receptor and may contribute to the genetic risk for addiction. Journal of neurochemistry. 2010;112:486–496. doi: 10.1111/j.1471-4159.2009.06472.x. [DOI] [PubMed] [Google Scholar]
- 17.Befort K, Filliol D, Decaillot FM, Gaveriaux-Ruff C, Hoehe MR, Kieffer BL. A single nucleotide polymorphic mutation in the human mu-opioid receptor severely impairs receptor signaling. J Biol Chem. 2001;276:3130–3137. doi: 10.1074/jbc.M006352200. [DOI] [PubMed] [Google Scholar]
- 18.Ray R, Ruparel K, Newberg A, Wileyto EP, Loughead JW, Divgi C, Blendy JA, Logan J, Zubieta JK, Lerman C. Human Mu Opioid Receptor (OPRM1 A118G) polymorphism is associated with brain mu-opioid receptor binding potential in smokers. Proc Natl Acad Sci U S A. 2011;108:9268–9273. doi: 10.1073/pnas.1018699108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Weerts EM, McCaul ME, Kuwabara H, Yang X, Xu X, Dannals RF, Frost JJ, Wong DF, Wand GS. Influence of OPRM1 Asn40Asp variant (A118G) on [11C]carfentanil binding potential: preliminary findings in human subjects. Int J Neuropsychopharmacol. 2013;16:47–53. doi: 10.1017/S146114571200017X. [DOI] [PubMed] [Google Scholar]
- 20.Zhang Y, Wang D, Johnson AD, Papp AC, Sadee W. Allelic expression imbalance of human mu opioid receptor (OPRM1) caused by variant A118G. J Biol Chem. 2005;280:32618–32624. doi: 10.1074/jbc.M504942200. [DOI] [PubMed] [Google Scholar]
- 21.Sillivan SE, Whittard JD, Jacobs MM, Ren Y, Mazloom AR, Caputi FF, Horvath M, Keller E, Ma’ayan A, Pan YX, Chiang LW, Hurd YL. ELK1 transcription factor linked to dysregulated striatal mu opioid receptor signaling network and OPRM1 polymorphism in human heroin abusers. Biological psychiatry. 2013;74:511–519. doi: 10.1016/j.biopsych.2013.04.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Oertel BG, Doehring A, Roskam B, Kettner M, Hackmann N, Ferreiros N, Schmidt PH, Lotsch J. Genetic-epigenetic interaction modulates mu-opioid receptor regulation. Hum Mol Genet. 2012;21:4751–4760. doi: 10.1093/hmg/dds314. [DOI] [PubMed] [Google Scholar]
- 23.Drakenberg K, Nikoshkov A, Horvath MC, Fagergren P, Gharibyan A, Saarelainen K, Rahman S, Nylander I, Bakalkin G, Rajs J, Keller E, Hurd YL. Mu Opioid receptor A118G polymorphism in association with striatal opioid neuropeptide gene expression in heroin abusers. Proc Natl Acad Sci U S A. 2006;103:7883–7888. doi: 10.1073/pnas.0600871103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Clarke TK, Crist RC, Kampman KM, Dackis CA, Pettinati HM, O’Brien CP, Oslin DW, Ferraro TN, Lohoff FW, Berrettini WH. Low frequency genetic variants in the mu-opioid receptor (OPRM1) affect risk for addiction to heroin and cocaine. Neurosci Lett. 2013;542:71–75. doi: 10.1016/j.neulet.2013.02.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Levran O, Londono D, O’Hara K, Nielsen DA, Peles E, Rotrosen J, Casadonte P, Linzy S, Randesi M, Ott J, Adelson M, Kreek MJ. Genetic susceptibility to heroin addiction: a candidate gene association study. Genes Brain Behav. 2008;7:720–729. doi: 10.1111/j.1601-183X.2008.00410.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Hancock DB, Levy JL, Gaddis NC, Glasheen C, Saccone NL, Page GP, Hulse GK, Wildenauer D, Kelty EA, Schwab SG, Degenhardt L, Martin NG, Montgomery GW, Attia J, Holliday EG, McEvoy M, Scott RJ, Bierut LJ, Nelson EC, Kral AH, Johnson EO. Cis-Expression Quantitative Trait Loci Mapping Reveals Replicable Associations with Heroin Addiction in OPRM1. Biological psychiatry. 2015;78:474–484. doi: 10.1016/j.biopsych.2015.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Nielsen DA, Ji F, Yuferov V, Ho A, Chen A, Levran O, Ott J, Kreek MJ. Genotype patterns that contribute to increased risk for or protection from developing heroin addiction. Mol Psychiatry. 2008;13:417–428. doi: 10.1038/sj.mp.4002147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Bart G, Heilig M, LaForge KS, Pollak L, Leal SM, Ott J, Kreek MJ. Substantial attributable risk related to a functional mu-opioid receptor gene polymorphism in association with heroin addiction in central Sweden. Mol Psychiatry. 2004;9:547–549. doi: 10.1038/sj.mp.4001504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Nikolov MA, Beltcheva O, Galabova A, Ljubenova A, Jankova E, Gergov G, Russev AA, Lynskey MT, Nelson EC, Nesheva E, Krasteva D, Lazarov P, Mitev VI, Kremensky IM, Kaneva RP, Todorov AA. No evidence of association between 118A>G OPRM1 polymorphism and heroin dependence in a large Bulgarian case-control sample. Drug Alcohol Depend. 2011;117:62–65. doi: 10.1016/j.drugalcdep.2010.12.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Woodcock EA, Lundahl LH, Burmeister M, Greenwald MK. Functional mu opioid receptor polymorphism (OPRM1 A(118) G) associated with heroin use outcomes in Caucasian males: A pilot study. Am J Addict. 2015;24:329–335. doi: 10.1111/ajad.12187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Coller JK, Beardsley J, Bignold J, Li Y, Merg F, Sullivan T, Cox TC, Somogyi AA. Lack of association between the A118G polymorphism of the mu opioid receptor gene (OPRM1) and opioid dependence: A meta-analysis. Pharmacogenomics and personalized medicine. 2009;2:9–19. [PMC free article] [PubMed] [Google Scholar]
- 32.Haerian BS, Haerian MS. OPRM1 rs1799971 polymorphism and opioid dependence: evidence from a meta-analysis. Pharmacogenomics. 2013;14:813–824. doi: 10.2217/pgs.13.57. [DOI] [PubMed] [Google Scholar]
- 33.Schwantes-An TH, Zhang J, Chen LS, Hartz SM, Culverhouse RC, Chen X, Coon H, Frank J, Kamens HM, Konte B, Kovanen L, Latvala A, Legrand LN, Maher BS, Melroy WE, Nelson EC, Reid MW, Robinson JD, Shen PH, Yang BZ, Andrews JA, Aveyard P, Beltcheva O, Brown SA, Cannon DS, Cichon S, Corley RP, Dahmen N, Degenhardt L, Foroud T, Gaebel W, Giegling I, Glatt SJ, Grucza RA, Hardin J, Hartmann AM, Heath AC, Herms S, Hodgkinson CA, Hoffmann P, Hops H, Huizinga D, Ising M, Johnson EO, Johnstone E, Kaneva RP, Kendler KS, Kiefer F, Kranzler HR, Krauter KS, Levran O, Lucae S, Lynskey MT, Maier W, Mann K, Martin NG, Mattheisen M, Montgomery GW, Muller-Myhsok B, Murphy MF, Neale MC, Nikolov MA, Nishita D, Nothen MM, Nurnberger J, Partonen T, Pergadia ML, Reynolds M, Ridinger M, Rose RJ, Rouvinen-Lagerstrom N, Scherbaum N, Schmal C, Soyka M, Stallings MC, Steffens M, Treutlein J, Tsuang M, Wall TL, Wodarz N, Yuferov V, Zill P, Bergen AW, Chen J, Cinciripini PM, Edenberg HJ, Ehringer MA, Ferrell RE, Gelernter J, Goldman D, Hewitt JK, Hopfer CJ, Iacono WG, Kaprio J, Kreek MJ, Kremensky IM, Madden PA, McGue M, Munafo MR, Philibert RA, Rietschel M, Roy A, Rujescu D, Saarikoski ST, Swan GE, Todorov AA, Vanyukov MM, Weiss RB, Bierut LJ, Saccone NL. Association of the OPRM1 Variant rs1799971 (A118G) with Non-Specific Liability to Substance Dependence in a Collaborative de novo Meta-Analysis of European-Ancestry Cohorts. Behav Genet. 2016;46:151–169. doi: 10.1007/s10519-015-9737-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Anton RF, Oroszi G, O’Malley S, Couper D, Swift R, Pettinati H, Goldman D. An evaluation of mu-opioid receptor (OPRM1) as a predictor of naltrexone response in the treatment of alcohol dependence: results from the Combined Pharmacotherapies and Behavioral Interventions for Alcohol Dependence (COMBINE) study. Arch Gen Psychiatry. 2008;65:135–144. doi: 10.1001/archpsyc.65.2.135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Oslin DW, Berrettini W, Kranzler HR, Pettinati H, Gelernter J, Volpicelli JR, O’Brien CP. A functional polymorphism of the mu-opioid receptor gene is associated with naltrexone response in alcohol-dependent patients. Neuropsychopharmacology. 2003;28:1546–1552. doi: 10.1038/sj.npp.1300219. [DOI] [PubMed] [Google Scholar]
- 36.Smith AH, Jensen KP, Li J, Nunez Y, Farrer LA, Hakonarson H, Cook-Sather SD, Kranzler HR, Gelernter J. Genome-wide association study of therapeutic opioid dosing identifies a novel locus upstream of OPRM1. Mol Psychiatry. 2017;22:346–352. doi: 10.1038/mp.2016.257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Hwang IC, Park JY, Myung SK, Ahn HY, Fukuda K, Liao Q. OPRM1 A118G gene variant and postoperative opioid requirement: a systematic review and meta-analysis. Anesthesiology. 2014;121:825–834. doi: 10.1097/ALN.0000000000000405. [DOI] [PubMed] [Google Scholar]
- 38.Hajj A, Halepian L, Osta NE, Chahine G, Kattan J, Rabbaa Khabbaz L. OPRM1 c.118A>G Polymorphism and Duration of Morphine Treatment Associated with Morphine Doses and Quality-of-Life in Palliative Cancer Pain Settings. Int J Mol Sci. 2017;18 doi: 10.3390/ijms18040669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Cajanus K, Kaunisto MA, Tallgren M, Jokela R, Kalso E. How much oxycodone is needed for adequate analgesia after breast cancer surgery: effect of the OPRM1 118A>G polymorphism. J Pain. 2014;15:1248–1256. doi: 10.1016/j.jpain.2014.09.002. [DOI] [PubMed] [Google Scholar]
- 40.Mouly S, Bloch V, Peoc’h K, Houze P, Labat L, Ksouda K, Simoneau G, Decleves X, Bergmann JF, Scherrmann JM, Laplanche JL, Lepine JP, Vorspan F. Methadone dose in heroin-dependent patients: role of clinical factors, comedications, genetic polymorphisms and enzyme activity. Br J Clin Pharmacol. 2015;79:967–977. doi: 10.1111/bcp.12576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Ahmad T, Valentovic MA, Rankin GO. Effects of cytochrome P450 single nucleotide polymorphisms on methadone metabolism and pharmacodynamics. Biochem Pharmacol. 2018 doi: 10.1016/j.bcp.2018.02.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Pan YX. Diversity and complexity of the mu opioid receptor gene: alternative pre-mRNA splicing and promoters. DNA Cell Biol. 2005;24:736–750. doi: 10.1089/dna.2005.24.736. [DOI] [PubMed] [Google Scholar]
- 43.Xu J, Lu Z, Xu M, Pan L, Deng Y, Xie X, Liu H, Ding S, Hurd YL, Pasternak GW, Klein RJ, Cartegni L, Zhou W, Pan YX. A heroin addiction severity-associated intronic single nucleotide polymorphism modulates alternative pre-mRNA splicing of the mu opioid receptor gene OPRM1 via hnRNPH interactions. J Neurosci. 2014;34:11048–11066. doi: 10.1523/JNEUROSCI.3986-13.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Xu J, Lu Z, Narayan A, Le Rouzic VP, Xu M, Hunkele A, Brown TG, Hoefer WF, Rossi GC, Rice RC, Martinez-Rivera A, Rajadhyaksha AM, Cartegni L, Bassoni DL, Pasternak GW, Pan YX. Alternatively spliced mu opioid receptor C termini impact the diverse actions of morphine. J Clin Invest. 2017;127:1561–1573. doi: 10.1172/JCI88760. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Pan YX, Xu J, Xu M, Rossi GC, Matulonis JE, Pasternak GW. Involvement of exon 11-associated variants of the mu opioid receptor MOR-1 in heroin, but not morphine, actions. Proc Natl Acad Sci U S A. 2009;106:4917–4922. doi: 10.1073/pnas.0811586106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Majumdar S, Grinnell S, Le Rouzic V, Burgman M, Polikar L, Ansonoff M, Pintar J, Pan YX, Pasternak GW. Truncated G protein-coupled mu opioid receptor MOR-1 splice variants are targets for highly potent opioid analgesics lacking side effects. Proc Natl Acad Sci U S A. 2011;108:19778–19783. doi: 10.1073/pnas.1115231108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Lu Z, Xu J, Rossi GC, Majumdar S, Pasternak GW, Pan YX. Mediation of opioid analgesia by a truncated 6-transmembrane GPCR. J Clin Invest. 2015;125:2626–2630. doi: 10.1172/JCI81070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Siuda ER, Carr R, 3rd, Rominger DH, Violin JD. Biased mu-opioid receptor ligands: a promising new generation of pain therapeutics. Curr Opin Pharmacol. 2017;32:77–84. doi: 10.1016/j.coph.2016.11.007. [DOI] [PubMed] [Google Scholar]
- 49.Winpenny D, Clark M, Cawkill D. Biased ligand quantification in drug discovery: from theory to high throughput screening to identify new biased mu opioid receptor agonists. Br J Pharmacol. 2016;173:1393–1403. doi: 10.1111/bph.13441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Violin JD, Crombie AL, Soergel DG, Lark MW. Biased ligands at G-protein-coupled receptors: promise and progress. Trends Pharmacol Sci. 2014;35:308–316. doi: 10.1016/j.tips.2014.04.007. [DOI] [PubMed] [Google Scholar]
- 51.Soergel DG, Subach RA, Burnham N, Lark MW, James IE, Sadler BM, Skobieranda F, Violin JD, Webster LR. Biased agonism of the mu-opioid receptor by TRV130 increases analgesia and reduces on-target adverse effects versus morphine: A randomized, double-blind, placebo-controlled, crossover study in healthy volunteers. Pain. 2014;155:1829–1835. doi: 10.1016/j.pain.2014.06.011. [DOI] [PubMed] [Google Scholar]
- 52.Schmid CL, Kennedy NM, Ross NC, Lovell KM, Yue Z, Morgenweck J, Cameron MD, Bannister TD, Bohn LM. Bias Factor and Therapeutic Window Correlate to Predict Safer Opioid Analgesics. Cell. 2017;171:1165–1175 e1113. doi: 10.1016/j.cell.2017.10.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Egervari G, Landry J, Callens J, Fullard JF, Roussos P, Keller E, Hurd YL. Striatal H3K27 Acetylation Linked to Glutamatergic Gene Dysregulation in Human Heroin Abusers Holds Promise as Therapeutic Target. Biological psychiatry. 2017;81:585–594. doi: 10.1016/j.biopsych.2016.09.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Nestler EJ. Epigenetic mechanisms of drug addiction. Neuropharmacology. 2014;76(Pt B):259–268. doi: 10.1016/j.neuropharm.2013.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Vaillancourt K, Ernst C, Mash D, Turecki G. DNA Methylation Dynamics and Cocaine in the Brain: Progress and Prospects. Genes (Basel) 2017;8 doi: 10.3390/genes8050138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Berkel TD, Pandey SC. Emerging Role of Epigenetic Mechanisms in Alcohol Addiction. Alcohol Clin Exp Res. 2017;41:666–680. doi: 10.1111/acer.13338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Viet CT, Dang D, Aouizerat BE, Miaskowski C, Ye Y, Viet DT, Ono K, Schmidt BL. OPRM1 Methylation Contributes to Opioid Tolerance in Cancer Patients. J Pain. 2017;18:1046–1059. doi: 10.1016/j.jpain.2017.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Ebrahimi G, Asadikaram G, Akbari H, Nematollahi MH, Abolhassani M, Shahabinejad G, Khodadadnejad L, Hashemi M. Elevated levels of DNA methylation at the OPRM1 promoter region in men with opioid use disorder. Am J Drug Alcohol Abuse. 2018;44:193–199. doi: 10.1080/00952990.2016.1275659. [DOI] [PubMed] [Google Scholar]
- 59.Doehring A, Oertel BG, Sittl R, Lotsch J. Chronic opioid use is associated with increased DNA methylation correlating with increased clinical pain. Pain. 2013;154:15–23. doi: 10.1016/j.pain.2012.06.011. [DOI] [PubMed] [Google Scholar]
- 60.Chorbov VM, Todorov AA, Lynskey MT, Cicero TJ. Elevated levels of DNA methylation at the OPRM1 promoter in blood and sperm from male opioid addicts. J Opioid Manag. 2011;7:258–264. doi: 10.5055/jom.2011.0067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Nielsen DA, Yuferov V, Hamon S, Jackson C, Ho A, Ott J, Kreek MJ. Increased OPRM1 DNA methylation in lymphocytes of methadone-maintained former heroin addicts. Neuropsychopharmacology. 2009;34:867–873. doi: 10.1038/npp.2008.108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Hwang CK, Kim CS, Kim DK, Law PY, Wei LN, Loh HH. Up-regulation of the mu-opioid receptor gene is mediated through chromatin remodeling and transcriptional factors in differentiated neuronal cells. Mol Pharmacol. 2010;78:58–68. doi: 10.1124/mol.110.064311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Oertel BG, Kettner M, Scholich K, Renne C, Roskam B, Geisslinger G, Schmidt PH, Lotsch J. A common human micro-opioid receptor genetic variant diminishes the receptor signaling efficacy in brain regions processing the sensory information of pain. J Biol Chem. 2009;284:6530–6535. doi: 10.1074/jbc.M807030200. [DOI] [PubMed] [Google Scholar]
- 64.Ferrer-Alcon M, La Harpe R, Garcia-Sevilla JA. Decreased immunodensities of micro-opioid receptors, receptor kinases GRK 2/6 and beta-arrestin-2 in postmortem brains of opiate addicts. Brain Res Mol Brain Res. 2004;121:114–122. doi: 10.1016/j.molbrainres.2003.11.009. [DOI] [PubMed] [Google Scholar]
- 65.Kozlenkov A, Jaffe AE, Timashpolsky A, Apontes P, Rudchenko S, Barbu M, Byne W, Hurd YL, Horvath S, Dracheva S. DNA Methylation Profiling of Human Prefrontal Cortex Neurons in Heroin Users Shows Significant Difference between Genomic Contexts of Hyper- and Hypomethylation and a Younger Epigenetic Age. Genes (Basel) 2017;8 doi: 10.3390/genes8060152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Allis CD, Jenuwein T. The molecular hallmarks of epigenetic control. Nat Rev Genet. 2016;17:487–500. doi: 10.1038/nrg.2016.59. [DOI] [PubMed] [Google Scholar]
- 67.Arrowsmith CH, Bountra C, Fish PV, Lee K, Schapira M. Epigenetic protein families: a new frontier for drug discovery. Nat Rev Drug Discov. 2012;11:384–400. doi: 10.1038/nrd3674. [DOI] [PubMed] [Google Scholar]
- 68.Bobadilla AC, Heinsbroek JA, Gipson CD, Griffin WC, Fowler CD, Kenny PJ, Kalivas PW. Corticostriatal plasticity, neuronal ensembles, and regulation of drug-seeking behavior. Prog Brain Res. 2017;235:93–112. doi: 10.1016/bs.pbr.2017.07.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Kalivas PW. The glutamate homeostasis hypothesis of addiction. Nat Rev Neurosci. 2009;10:561–572. doi: 10.1038/nrn2515. [DOI] [PubMed] [Google Scholar]
- 70.Bradford AC, Bradford WD. Medical Marijuana Laws: The Authors Reply. Health Aff (Millwood) 2016;35:1937. doi: 10.1377/hlthaff.2016.1110. [DOI] [PubMed] [Google Scholar]
- 71.Kim JH, Santaella-Tenorio J, Mauro C, Wrobel J, Cerda M, Keyes KM, Hasin D, Martins SS, Li G. State Medical Marijuana Laws and the Prevalence of Opioids Detected Among Fatally Injured Drivers. Am J Public Health. 2016;106:2032–2037. doi: 10.2105/AJPH.2016.303426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Santaella-Tenorio J, Mauro CM, Wall MM, Kim JH, Cerda M, Keyes KM, Hasin DS, Galea S, Martins SS. US Traffic Fatalities, 1985-2014, and Their Relationship to Medical Marijuana Laws. Am J Public Health. 2017;107:336–342. doi: 10.2105/AJPH.2016.303577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Powell D, Pacula RL, Jacobson M. Do medical marijuana laws reduce addictions and deaths related to pain killers? J Health Econ. 2018;58:29–42. doi: 10.1016/j.jhealeco.2017.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Shi Y. Medical marijuana policies and hospitalizations related to marijuana and opioid pain reliever. Drug Alcohol Depend. 2017;173:144–150. doi: 10.1016/j.drugalcdep.2017.01.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Stopponi S, Soverchia L, Ubaldi M, Cippitelli A, Serpelloni G, Ciccocioppo R. Chronic THC during adolescence increases the vulnerability to stress-induced relapse to heroin seeking in adult rats. European neuropsychopharmacology : the journal of the European College of Neuropsychopharmacology. 2014;24:1037–1045. doi: 10.1016/j.euroneuro.2013.12.012. [DOI] [PubMed] [Google Scholar]
- 76.Cadoni C, Simola N, Espa E, Fenu S, Di Chiara G. Strain dependence of adolescent Cannabis influence on heroin reward and mesolimbic dopamine transmission in adult Lewis and Fischer 344 rats. Addiction biology. 2015;20:132–142. doi: 10.1111/adb.12085. [DOI] [PubMed] [Google Scholar]
- 77.Tomasiewicz HC, Jacobs MM, Wilkinson MB, Wilson SP, Nestler EJ, Hurd YL. Proenkephalin mediates the enduring effects of adolescent cannabis exposure associated with adult opiate vulnerability. Biological psychiatry. 2012;72:803–810. doi: 10.1016/j.biopsych.2012.04.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Ellgren M, Spano SM, Hurd YL. Adolescent cannabis exposure alters opiate intake and opioid limbic neuronal populations in adult rats. Neuropsychopharmacology. 2007;32:607–615. doi: 10.1038/sj.npp.1301127. [DOI] [PubMed] [Google Scholar]
- 79.Katsidoni V, Anagnostou I, Panagis G. Cannabidiol inhibits the reward-facilitating effect of morphine: involvement of 5-HT1A receptors in the dorsal raphe nucleus. Addiction biology. 2013;18:286–296. doi: 10.1111/j.1369-1600.2012.00483.x. [DOI] [PubMed] [Google Scholar]
- 80.Ren Y, Whittard J, Higuera-Matas A, Morris CV, Hurd YL. Cannabidiol, a nonpsychotropic component of cannabis, inhibits cue-induced heroin seeking and normalizes discrete mesolimbic neuronal disturbances. J Neurosci. 2009;29:14764–14769. doi: 10.1523/JNEUROSCI.4291-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Hurd YL, Yoon M, Manini AF, Hernandez S, Olmedo R, Ostman M, Jutras-Aswad D. Early Phase in the Development of Cannabidiol as a Treatment for Addiction: Opioid Relapse Takes Initial Center Stage. Neurotherapeutics. 2015;12:807–815. doi: 10.1007/s13311-015-0373-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Parker LA, Burton P, Sorge RE, Yakiwchuk C, Mechoulam R. Effect of low doses of delta9-tetrahydrocannabinol and cannabidiol on the extinction of cocaine-induced and amphetamine-induced conditioned place preference learning in rats. Psychopharmacology (Berl) 2004;175:360–366. doi: 10.1007/s00213-004-1825-7. [DOI] [PubMed] [Google Scholar]
- 83.Vann RE, Gamage TF, Warner JA, Marshall EM, Taylor NL, Martin BR, Wiley JL. Divergent effects of cannabidiol on the discriminative stimulus and place conditioning effects of Delta(9)-tetrahydrocannabinol. Drug Alcohol Depend. 2008;94:191–198. doi: 10.1016/j.drugalcdep.2007.11.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Babalonis S, Haney M, Malcolm RJ, Lofwall MR, Votaw VR, Sparenborg S, Walsh SL. Oral cannabidiol does not produce a signal for abuse liability in frequent marijuana smokers. Drug Alcohol Depend. 2017;172:9–13. doi: 10.1016/j.drugalcdep.2016.11.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Haney M, Malcolm RJ, Babalonis S, Nuzzo PA, Cooper ZD, Bedi G, Gray KM, McRae-Clark A, Lofwall MR, Sparenborg S, Walsh SL. Oral Cannabidiol does not Alter the Subjective, Reinforcing or Cardiovascular Effects of Smoked Cannabis. Neuropsychopharmacology. 2016;41:1974–1982. doi: 10.1038/npp.2015.367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Manini AF, Yiannoulos G, Bergamaschi MM, Hernandez S, Olmedo R, Barnes AJ, Winkel G, Sinha R, Jutras-Aswad D, Huestis MA, Hurd YL. Safety and pharmacokinetics of oral cannabidiol when administered concomitantly with intravenous fentanyl in humans. J Addict Med. 2015;9:204–210. doi: 10.1097/ADM.0000000000000118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.McGuire P, Robson P, Cubala WJ, Vasile D, Morrison PD, Barron R, Taylor A, Wright S. Cannabidiol (CBD) as an Adjunctive Therapy in Schizophrenia: A Multicenter Randomized Controlled Trial. Am J Psychiatry. 2018;175:225–231. doi: 10.1176/appi.ajp.2017.17030325. [DOI] [PubMed] [Google Scholar]
- 88.Iffland K, Grotenhermen F. An Update on Safety and Side Effects of Cannabidiol: A Review of Clinical Data and Relevant Animal Studies. Cannabis Cannabinoid Res. 2017;2:139–154. doi: 10.1089/can.2016.0034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Bonese KF, Wainer BH, Fitch FW, Rothberg RM, Schuster CR. Changes in heroin self-administration by a rhesus monkey after morphine immunisation. Nature. 1974;252:708–710. doi: 10.1038/252708a0. [DOI] [PubMed] [Google Scholar]
- 90.Hatsukami DK, Jorenby DE, Gonzales D, Rigotti NA, Glover ED, Oncken CA, Tashkin DP, Reus VI, Akhavain RC, Fahim RE, Kessler PD, Niknian M, Kalnik MW, Rennard SI. Immunogenicity and smoking-cessation outcomes for a novel nicotine immunotherapeutic. Clin Pharmacol Ther. 2011;89:392–399. doi: 10.1038/clpt.2010.317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Wagena EJ, de Vos A, Horwith G, van Schayck CP. The immunogenicity and safety of a nicotine vaccine in smokers and nonsmokers: results of a randomized, placebo-controlled phase 1/2 trial. Nicotine Tob Res. 2008;10:213–218. doi: 10.1080/14622200701704921. [DOI] [PubMed] [Google Scholar]
- 92.Hartmann-Boyce J, Cahill K, Hatsukami D, Cornuz J. Nicotine vaccines for smoking cessation. Cochrane Database Syst Rev. 2012:CD007072. doi: 10.1002/14651858.CD007072.pub2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Haney M, Gunderson EW, Jiang H, Collins ED, Foltin RW. Cocaine-specific antibodies blunt the subjective effects of smoked cocaine in humans. Biological psychiatry. 2010;67:59–65. doi: 10.1016/j.biopsych.2009.08.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Martell BA, Orson FM, Poling J, Mitchell E, Rossen RD, Gardner T, Kosten TR. Cocaine vaccine for the treatment of cocaine dependence in methadone-maintained patients: a randomized, double-blind, placebo-controlled efficacy trial. Arch Gen Psychiatry. 2009;66:1116–1123. doi: 10.1001/archgenpsychiatry.2009.128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Kosten TR, Rosen M, Bond J, Settles M, Roberts JS, Shields J, Jack L, Fox B. Human therapeutic cocaine vaccine: safety and immunogenicity. Vaccine. 2002;20:1196–1204. doi: 10.1016/s0264-410x(01)00425-x. [DOI] [PubMed] [Google Scholar]
- 96.Kosten TR, Domingo CB, Shorter D, Orson F, Green C, Somoza E, Sekerka R, Levin FR, Mariani JJ, Stitzer M, Tompkins DA, Rotrosen J, Thakkar V, Smoak B, Kampman K. Vaccine for cocaine dependence: a randomized double-blind placebo-controlled efficacy trial. Drug Alcohol Depend. 2014;140:42–47. doi: 10.1016/j.drugalcdep.2014.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Hwang CS, Bremer PT, Wenthur CJ, Ho SO, Chiang S, Ellis B, Zhou B, Fujii G, Janda KD. Enhancing Efficacy and Stability of an Antiheroin Vaccine: Examination of Antinociception, Opioid Binding Profile, and Lethality. Mol Pharm. 2018;15:1062–1072. doi: 10.1021/acs.molpharmaceut.7b00933. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Bremer PT, Janda KD. Conjugate Vaccine Immunotherapy for Substance Use Disorder. Pharmacol Rev. 2017;69:298–315. doi: 10.1124/pr.117.013904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Bremer PT, Schlosburg JE, Banks ML, Steele FF, Zhou B, Poklis JL, Janda KD. Development of a Clinically Viable Heroin Vaccine. J Am Chem Soc. 2017;139:8601–8611. doi: 10.1021/jacs.7b03334. [DOI] [PMC free article] [PubMed] [Google Scholar]