Change Password

Your password must have 6 characters or more:.

  • a lower case character, 
  • an upper case character, 
  • a special character 

Password Changed Successfully

Your password has been changed

Create your account

Forget yout password.

Enter your email address below and we will send you the reset instructions

If the address matches an existing account you will receive an email with instructions to reset your password

Forgot your Username?

Enter your email address below and we will send you your username

If the address matches an existing account you will receive an email with instructions to retrieve your username

Psychiatry Online

  • April 01, 2024 | VOL. 181, NO. 4 CURRENT ISSUE pp.255-346
  • March 01, 2024 | VOL. 181, NO. 3 pp.171-254
  • February 01, 2024 | VOL. 181, NO. 2 pp.83-170
  • January 01, 2024 | VOL. 181, NO. 1 pp.1-82

The American Psychiatric Association (APA) has updated its Privacy Policy and Terms of Use , including with new information specifically addressed to individuals in the European Economic Area. As described in the Privacy Policy and Terms of Use, this website utilizes cookies, including for the purpose of offering an optimal online experience and services tailored to your preferences.

Please read the entire Privacy Policy and Terms of Use. By closing this message, browsing this website, continuing the navigation, or otherwise continuing to use the APA's websites, you confirm that you understand and accept the terms of the Privacy Policy and Terms of Use, including the utilization of cookies.

Substance Use Disorders and Addiction: Mechanisms, Trends, and Treatment Implications

  • Ned H. Kalin , M.D.

Search for more papers by this author

The numbers for substance use disorders are large, and we need to pay attention to them. Data from the 2018 National Survey on Drug Use and Health ( 1 ) suggest that, over the preceding year, 20.3 million people age 12 or older had substance use disorders, and 14.8 million of these cases were attributed to alcohol. When considering other substances, the report estimated that 4.4 million individuals had a marijuana use disorder and that 2 million people suffered from an opiate use disorder. It is well known that stress is associated with an increase in the use of alcohol and other substances, and this is particularly relevant today in relation to the chronic uncertainty and distress associated with the COVID-19 pandemic along with the traumatic effects of racism and social injustice. In part related to stress, substance use disorders are highly comorbid with other psychiatric illnesses: 9.2 million adults were estimated to have a 1-year prevalence of both a mental illness and at least one substance use disorder. Although they may not necessarily meet criteria for a substance use disorder, it is well known that psychiatric patients have increased usage of alcohol, cigarettes, and other illicit substances. As an example, the survey estimated that over the preceding month, 37.2% of individuals with serious mental illnesses were cigarette smokers, compared with 16.3% of individuals without mental illnesses. Substance use frequently accompanies suicide and suicide attempts, and substance use disorders are associated with a long-term increased risk of suicide.

Addiction is the key process that underlies substance use disorders, and research using animal models and humans has revealed important insights into the neural circuits and molecules that mediate addiction. More specifically, research has shed light onto mechanisms underlying the critical components of addiction and relapse: reinforcement and reward, tolerance, withdrawal, negative affect, craving, and stress sensitization. In addition, clinical research has been instrumental in developing an evidence base for the use of pharmacological agents in the treatment of substance use disorders, which, in combination with psychosocial approaches, can provide effective treatments. However, despite the existence of therapeutic tools, relapse is common, and substance use disorders remain grossly undertreated. For example, whether at an inpatient hospital treatment facility or at a drug or alcohol rehabilitation program, it was estimated that only 11% of individuals needing treatment for substance use received appropriate care in 2018. Additionally, it is worth emphasizing that current practice frequently does not effectively integrate dual diagnosis treatment approaches, which is important because psychiatric and substance use disorders are highly comorbid. The barriers to receiving treatment are numerous and directly interact with existing health care inequities. It is imperative that as a field we overcome the obstacles to treatment, including the lack of resources at the individual level, a dearth of trained providers and appropriate treatment facilities, racial biases, and the marked stigmatization that is focused on individuals with addictions.

This issue of the Journal is focused on understanding factors contributing to substance use disorders and their comorbidity with psychiatric disorders, the effects of prenatal alcohol use on preadolescents, and brain mechanisms that are associated with addiction and relapse. An important theme that emerges from this issue is the necessity for understanding maladaptive substance use and its treatment in relation to health care inequities. This highlights the imperative to focus resources and treatment efforts on underprivileged and marginalized populations. The centerpiece of this issue is an overview on addiction written by Dr. George Koob, the director of the National Institute on Alcohol Abuse and Alcoholism (NIAAA), and coauthors Drs. Patricia Powell (NIAAA deputy director) and Aaron White ( 2 ). This outstanding article will serve as a foundational knowledge base for those interested in understanding the complex factors that mediate drug addiction. Of particular interest to the practice of psychiatry is the emphasis on the negative affect state “hyperkatifeia” as a major driver of addictive behavior and relapse. This places the dysphoria and psychological distress that are associated with prolonged withdrawal at the heart of treatment and underscores the importance of treating not only maladaptive drug-related behaviors but also the prolonged dysphoria and negative affect associated with addiction. It also speaks to why it is crucial to concurrently treat psychiatric comorbidities that commonly accompany substance use disorders.

Insights Into Mechanisms Related to Cocaine Addiction Using a Novel Imaging Method for Dopamine Neurons

Cassidy et al. ( 3 ) introduce a relatively new imaging technique that allows for an estimation of dopamine integrity and function in the substantia nigra, the site of origin of dopamine neurons that project to the striatum. Capitalizing on the high levels of neuromelanin that are found in substantia nigra dopamine neurons and the interaction between neuromelanin and intracellular iron, this MRI technique, termed neuromelanin-sensitive MRI (NM-MRI), shows promise in studying the involvement of substantia nigra dopamine neurons in neurodegenerative diseases and psychiatric illnesses. The authors used this technique to assess dopamine function in active cocaine users with the aim of exploring the hypothesis that cocaine use disorder is associated with blunted presynaptic striatal dopamine function that would be reflected in decreased “integrity” of the substantia nigra dopamine system. Surprisingly, NM-MRI revealed evidence for increased dopamine in the substantia nigra of individuals using cocaine. The authors suggest that this finding, in conjunction with prior work suggesting a blunted dopamine response, points to the possibility that cocaine use is associated with an altered intracellular distribution of dopamine. Specifically, the idea is that dopamine is shifted from being concentrated in releasable, functional vesicles at the synapse to a nonreleasable cytosolic pool. In addition to providing an intriguing alternative hypothesis underlying the cocaine-related alterations observed in substantia nigra dopamine function, this article highlights an innovative imaging method that can be used in further investigations involving the role of substantia nigra dopamine systems in neuropsychiatric disorders. Dr. Charles Bradberry, chief of the Preclinical Pharmacology Section at the National Institute on Drug Abuse, contributes an editorial that further explains the use of NM-MRI and discusses the theoretical implications of these unexpected findings in relation to cocaine use ( 4 ).

Treatment Implications of Understanding Brain Function During Early Abstinence in Patients With Alcohol Use Disorder

Developing a better understanding of the neural processes that are associated with substance use disorders is critical for conceptualizing improved treatment approaches. Blaine et al. ( 5 ) present neuroimaging data collected during early abstinence in patients with alcohol use disorder and link these data to relapses occurring during treatment. Of note, the findings from this study dovetail with the neural circuit schema Koob et al. provide in this issue’s overview on addiction ( 2 ). The first study in the Blaine et al. article uses 44 patients and 43 control subjects to demonstrate that patients with alcohol use disorder have a blunted neural response to the presentation of stress- and alcohol-related cues. This blunting was observed mainly in the ventromedial prefrontal cortex, a key prefrontal regulatory region, as well as in subcortical regions associated with reward processing, specifically the ventral striatum. Importantly, this finding was replicated in a second study in which 69 patients were studied in relation to their length of abstinence prior to treatment and treatment outcomes. The results demonstrated that individuals with the shortest abstinence times had greater alterations in neural responses to stress and alcohol cues. The authors also found that an individual’s length of abstinence prior to treatment, independent of the number of days of abstinence, was a predictor of relapse and that the magnitude of an individual’s neural alterations predicted the amount of heavy drinking occurring early in treatment. Although relapse is an all too common outcome in patients with substance use disorders, this study highlights an approach that has the potential to refine and develop new treatments that are based on addiction- and abstinence-related brain changes. In her thoughtful editorial, Dr. Edith Sullivan from Stanford University comments on the details of the study, the value of studying patients during early abstinence, and the implications of these findings for new treatment development ( 6 ).

Relatively Low Amounts of Alcohol Intake During Pregnancy Are Associated With Subtle Neurodevelopmental Effects in Preadolescent Offspring

Excessive substance use not only affects the user and their immediate family but also has transgenerational effects that can be mediated in utero. Lees et al. ( 7 ) present data suggesting that even the consumption of relatively low amounts of alcohol by expectant mothers can affect brain development, cognition, and emotion in their offspring. The researchers used data from the Adolescent Brain Cognitive Development Study, a large national community-based study, which allowed them to assess brain structure and function as well as behavioral, cognitive, and psychological outcomes in 9,719 preadolescents. The mothers of 2,518 of the subjects in this study reported some alcohol use during pregnancy, albeit at relatively low levels (0 to 80 drinks throughout pregnancy). Interestingly, and opposite of that expected in relation to data from individuals with fetal alcohol spectrum disorders, increases in brain volume and surface area were found in offspring of mothers who consumed the relatively low amounts of alcohol. Notably, any prenatal alcohol exposure was associated with small but significant increases in psychological problems that included increases in separation anxiety disorder and oppositional defiant disorder. Additionally, a dose-response effect was found for internalizing psychopathology, somatic complaints, and attentional deficits. While subtle, these findings point to neurodevelopmental alterations that may be mediated by even small amounts of prenatal alcohol consumption. Drs. Clare McCormack and Catherine Monk from Columbia University contribute an editorial that provides an in-depth assessment of these findings in relation to other studies, including those assessing severe deficits in individuals with fetal alcohol syndrome ( 8 ). McCormack and Monk emphasize that the behavioral and psychological effects reported in the Lees et al. article would not be clinically meaningful. However, it is feasible that the influences of these low amounts of alcohol could interact with other predisposing factors that might lead to more substantial negative outcomes.

Increased Comorbidity Between Substance Use and Psychiatric Disorders in Sexual Identity Minorities

There is no question that victims of societal marginalization experience disproportionate adversity and stress. Evans-Polce et al. ( 9 ) focus on this concern in relation to individuals who identify as sexual minorities by comparing their incidence of comorbid substance use and psychiatric disorders with that of individuals who identify as heterosexual. By using 2012−2013 data from 36,309 participants in the National Epidemiologic Study on Alcohol and Related Conditions–III, the authors examine the incidence of comorbid alcohol and tobacco use disorders with anxiety, mood disorders, and posttraumatic stress disorder (PTSD). The findings demonstrate increased incidences of substance use and psychiatric disorders in individuals who identified as bisexual or as gay or lesbian compared with those who identified as heterosexual. For example, a fourfold increase in the prevalence of PTSD was found in bisexual individuals compared with heterosexual individuals. In addition, the authors found an increased prevalence of substance use and psychiatric comorbidities in individuals who identified as bisexual and as gay or lesbian compared with individuals who identified as heterosexual. This was most prominent in women who identified as bisexual. For example, of the bisexual women who had an alcohol use disorder, 60.5% also had a psychiatric comorbidity, compared with 44.6% of heterosexual women. Additionally, the amount of reported sexual orientation discrimination and number of lifetime stressful events were associated with a greater likelihood of having comorbid substance use and psychiatric disorders. These findings are important but not surprising, as sexual minority individuals have a history of increased early-life trauma and throughout their lives may experience the painful and unwarranted consequences of bias and denigration. Nonetheless, these findings underscore the strong negative societal impacts experienced by minority groups and should sensitize providers to the additional needs of these individuals.

Trends in Nicotine Use and Dependence From 2001–2002 to 2012–2013

Although considerable efforts over earlier years have curbed the use of tobacco and nicotine, the use of these substances continues to be a significant public health problem. As noted above, individuals with psychiatric disorders are particularly vulnerable. Grant et al. ( 10 ) use data from the National Epidemiologic Survey on Alcohol and Related Conditions collected from a very large cohort to characterize trends in nicotine use and dependence over time. Results from their analysis support the so-called hardening hypothesis, which posits that although intervention-related reductions in nicotine use may have occurred over time, the impact of these interventions is less potent in individuals with more severe addictive behavior (i.e., nicotine dependence). When adjusted for sociodemographic factors, the results demonstrated a small but significant increase in nicotine use from 2001–2002 to 2012–2013. However, a much greater increase in nicotine dependence (46.1% to 52%) was observed over this time frame in individuals who had used nicotine during the preceding 12 months. The increases in nicotine use and dependence were associated with factors related to socioeconomic status, such as lower income and lower educational attainment. The authors interpret these findings as evidence for the hardening hypothesis, suggesting that despite the impression that nicotine use has plateaued, there is a growing number of highly dependent nicotine users who would benefit from nicotine dependence intervention programs. Dr. Kathleen Brady, from the Medical University of South Carolina, provides an editorial ( 11 ) that reviews the consequences of tobacco use and the history of the public measures that were initially taken to combat its use. Importantly, her editorial emphasizes the need to address health care inequity issues that affect individuals of lower socioeconomic status by devoting resources to develop and deploy effective smoking cessation interventions for at-risk and underresourced populations.

Conclusions

Maladaptive substance use and substance use disorders are highly prevalent and are among the most significant public health problems. Substance use is commonly comorbid with psychiatric disorders, and treatment efforts need to concurrently address both. The papers in this issue highlight new findings that are directly relevant to understanding, treating, and developing policies to better serve those afflicted with addictions. While treatments exist, the need for more effective treatments is clear, especially those focused on decreasing relapse rates. The negative affective state, hyperkatifeia, that accompanies longer-term abstinence is an important treatment target that should be emphasized in current practice as well as in new treatment development. In addition to developing a better understanding of the neurobiology of addictions and abstinence, it is necessary to ensure that there is equitable access to currently available treatments and treatment programs. Additional resources must be allocated to this cause. This depends on the recognition that health care inequities and societal barriers are major contributors to the continued high prevalence of substance use disorders, the individual suffering they inflict, and the huge toll that they incur at a societal level.

Disclosures of Editors’ financial relationships appear in the April 2020 issue of the Journal .

1 US Department of Health and Human Services: Substance Abuse and Mental Health Services Administration, Center for Behavioral Health Statistics and Quality: National Survey on Drug Use and Health 2018. Rockville, Md, SAMHSA, 2019 ( https://www.samhsa.gov/data/nsduh/reports-detailed-tables-2018-NSDUH ) Google Scholar

2 Koob GF, Powell P, White A : Addiction as a coping response: hyperkatifeia, deaths of despair, and COVID-19 . Am J Psychiatry 2020 ; 177:1031–1037 Link ,  Google Scholar

3 Cassidy CM, Carpenter KM, Konova AB, et al. : Evidence for dopamine abnormalities in the substantia nigra in cocaine addiction revealed by neuromelanin-sensitive MRI . Am J Psychiatry 2020 ; 177:1038–1047 Link ,  Google Scholar

4 Bradberry CW : Neuromelanin MRI: dark substance shines a light on dopamine dysfunction and cocaine use (editorial). Am J Psychiatry 2020 ; 177:1019–1021 Abstract ,  Google Scholar

5 Blaine SK, Wemm S, Fogelman N, et al. : Association of prefrontal-striatal functional pathology with alcohol abstinence days at treatment initiation and heavy drinking after treatment initiation . Am J Psychiatry 2020 ; 177:1048–1059 Abstract ,  Google Scholar

6 Sullivan EV : Why timing matters in alcohol use disorder recovery (editorial). Am J Psychiatry 2020 ; 177:1022–1024 Abstract ,  Google Scholar

7 Lees B, Mewton L, Jacobus J, et al. : Association of prenatal alcohol exposure with psychological, behavioral, and neurodevelopmental outcomes in children from the Adolescent Brain Cognitive Development Study . Am J Psychiatry 2020 ; 177:1060–1072 Link ,  Google Scholar

8 McCormack C, Monk C : Considering prenatal alcohol exposure in a developmental origins of health and disease framework (editorial). Am J Psychiatry 2020 ; 177:1025–1028 Abstract ,  Google Scholar

9 Evans-Polce RJ, Kcomt L, Veliz PT, et al. : Alcohol, tobacco, and comorbid psychiatric disorders and associations with sexual identity and stress-related correlates . Am J Psychiatry 2020 ; 177:1073–1081 Abstract ,  Google Scholar

10 Grant BF, Shmulewitz D, Compton WM : Nicotine use and DSM-IV nicotine dependence in the United States, 2001–2002 and 2012–2013 . Am J Psychiatry 2020 ; 177:1082–1090 Link ,  Google Scholar

11 Brady KT : Social determinants of health and smoking cessation: a challenge (editorial). Am J Psychiatry 2020 ; 177:1029–1030 Abstract ,  Google Scholar

  • Cited by None

drug addiction research topics

  • Substance-Related and Addictive Disorders
  • Addiction Psychiatry
  • Transgender (LGBT) Issues

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • View all journals
  • My Account Login
  • Explore content
  • About the journal
  • Publish with us
  • Sign up for alerts
  • Review Article
  • Open access
  • Published: 12 August 2021

Drug addiction: from bench to bedside

  • Julian Cheron 1 &
  • Alban de Kerchove d’Exaerde   ORCID: orcid.org/0000-0002-0682-5877 1  

Translational Psychiatry volume  11 , Article number:  424 ( 2021 ) Cite this article

14k Accesses

23 Citations

1 Altmetric

Metrics details

  • Molecular neuroscience

Drug addiction is responsible for millions of deaths per year around the world. Still, its management as a chronic disease is shadowed by misconceptions from the general public. Indeed, drug consumers are often labelled as “weak”, “immoral” or “depraved”. Consequently, drug addiction is often perceived as an individual problem and not societal. In technical terms, drug addiction is defined as a chronic, relapsing disease resulting from sustained effects of drugs on the brain. Through a better characterisation of the cerebral circuits involved, and the long-term modifications of the brain induced by addictive drugs administrations, first, we might be able to change the way the general public see the patient who is suffering from drug addiction, and second, we might be able to find new treatments to normalise the altered brain homeostasis. In this review, we synthetise the contribution of fundamental research to the understanding drug addiction and its contribution to potential novel therapeutics. Mostly based on drug-induced modifications of synaptic plasticity and epigenetic mechanisms (and their behavioural correlates) and after demonstration of their reversibility, we tried to highlight promising therapeutics. We also underline the specific temporal dynamics and psychosocial aspects of this complex psychiatric disease adding parameters to be considered in clinical trials and paving the way to test new therapeutic venues.

Similar content being viewed by others

drug addiction research topics

Addiction as a brain disease revised: why it still matters, and the need for consilience

Markus Heilig, James MacKillop, … Louk J. M. J. Vanderschuren

drug addiction research topics

The neurobiology of drug addiction: cross-species insights into the dysfunction and recovery of the prefrontal cortex

Ahmet O. Ceceli, Charles W. Bradberry & Rita Z. Goldstein

drug addiction research topics

Mechanisms and molecular targets surrounding the potential therapeutic effects of psychedelics

Alaina M. Jaster & Javier González-Maeso

Introduction

Drug addiction including smoking, alcohol and illicit drug use is indirectly or directly responsible for 11.8 million deaths each year in the world [ 1 ]. According to the Global Burden of Disease study, this number is higher than deaths from cancer and accounts for a fifth of all deaths around the world [ 1 ].

Drug addiction is defined as a chronic, relapsing disease that results from the prolonged effects of drugs on the brain. Similarly to other neuropsychiatric diseases, drug addiction is intermingled with behavioural and social aspects that are equally important parts of the disease, complicating the overall therapeutic approach. Actually, it is only recently, in the beginning of the 21st century, that we started to see “the drug-addict” as someone suffering from a disease and whose brain has been altered fundamentally by drugs [ 2 ]. Therefore, the most effective treatment approaches include biological, behavioural and social-context components. Based on the latest scientific advances, treatment and management of drug addiction patients point towards a personalised strategy [ 3 ]. However, there are very few objective and effective strategies for treating drug addiction. Without the mandatory mechanistic basic knowledge on drug addiction, the development of new therapeutic strategies is postponed.

The neurobiological circuits and mechanisms that support compulsive seeking and consumption of drugs with addictive potential are partially known. They comprise a progressive shift in the involvement of ventral to dorsal and medial to lateral striatal circuitry [ 4 , 5 ], along with molecular and cellular adaptations to drugs of abuse exposure. They include neuronal and synaptic plasticity and modifications in gene expression, in part through epigenetic mechanisms [ 6 ]. Notably, drug-induced neuronal modifications can also occur in non-pathological processes, underlying the fact that drugs of abuse hijack normal adaptive changes in the brain [ 7 ]. Indeed, laboratory and clinical observations suggest that addiction is driven by the usurpation of neuronal processes that normally serve reward-related learning and memory. Most of the modifications that have been shown to be involved in a state of addiction (modified gene transcription, epigenetics, neuronal plasticity and neurotrophic mechanisms) are also associated with physiological forms of behavioural memory in murine model such as spatial memory, fear conditioning and operant conditioning [ 7 , 8 ].

We know that only a proportion of individuals (depending of the drug type) will develop drug addiction after several exposures [ 9 ]. This individual vulnerability is probably linked to both genetic and environmental factors [ 10 ]. Drug addiction is highly polygenic, as hundreds of genetic variations combined result in variable vulnerability [ 11 , 12 ]. Several types of environmental factors have been characterised and interact with an individual genetic background [ 12 , 13 ]. Psychosocial stress is one of the factors, but the most important one, is by far, the exposure to drugs of abuse. Usually, drug abuse starts with a ‘gateway’ drug (mostly socially driven) catapulting the individual vulnerability to other drugs of abuse [ 14 ].

During the last three decades, combine effort has been dedicated to identify brain regions and molecular pathways involved in the development of addiction to drugs of abuse. Here, we will focus on experimental approaches that helped to provide a clearer picture on the physiopathology of drug addiction guiding therapeutic opportunities.

Converging actions on brain reward pathway elicit its remodelling

The circuit at the centre of the disease is the mesolimbic pathway, also referred as the reward pathway (Fig. 1 ). The mesolimbic pathway includes dopaminergic neurons in the ventral tegmental area (VTA) of the midbrain and their targets in the limbic forebrain, especially the nucleus accumbens (NAc), a major component of the ventral striatum. The GABA medium-sized spiny neurons (MSNs, ~95% of striatal neurons), which are targets of glutamatergic and dopaminergic inputs, form two pathways [ 15 ]. The dopamine D1 receptor–positive (D1R) striatonigral MSNs project to the medial globus pallidus and substantia nigra pars reticulata (direct pathway) and coexpress dopamine D1 receptors and substance P, whereas D2R striatopallidal MSNs project to the lateral globus pallidus (indirect pathway) and coexpress dopamine D2 receptor, adenosine A2A receptor and enkephalin [ 16 , 17 ]. Through different initial mechanisms, drugs of abuse increase the release of dopamine in the NAc from the VTA [ 18 , 19 , 20 ]. This VTA-NAc pathway could be seen as primum movens for the acute rewarding effects of all drugs of abuse. Regardless that drugs of abuse have distinct protein targets and mechanisms of action, in the end, the main addiction-related modifications are common to nearly all drugs of abuse and converge on the VTA and NAc with common acute functional effects [ 21 ]. It is schematically conjectured, that when stimulated by dopamine, cells in the NAc produce feelings of reward and satisfaction [ 22 ]. The physiological function of this response is to facilitate the motivation for basic biological goal-directed behaviours as survival, social interaction and reproduction. By artificially causing a build-up of dopamine in the NAc, drugs of abuse generate an artificial reward effect [ 22 ]. As all drugs of abuse increase dopaminergic transmission to the NAc after acute administration, they also produce shared modifications in the mesolimbic system after chronic exposure. They include (i) hypofunction of the dopamine pathway that is seen as a major contributor to the negative emotional symptoms associated to drug withdrawal, leading to drug craving and relapse, and (ii) drug-induced adaptations in glutamatergic afferents to the NAc [ 23 , 24 ]. Clearly, these modifications in the mesolimbic system after the exposure to drugs of abuse is oversimplified. The hypofunction of dopaminergic system hypothesis is self-fulfilling in that research work has principally focused on dopamine to the exclusion of other neurotransmitters. Actually, some drug of abuse reinforcement appears to be independent of the mesocorticolimbic dopamine system (e.g. opioids [ 25 ], nicotine [ 26 ]), but support self-administration by imitating the effect of dopamine in the nucleus accumbens [ 21 , 27 , 28 ].

figure 1

Addictive drugs of different types have a common effect of increasing levels of dopamine released by neurons projecting from the ventral tegmental area (VTA). This effect is central for initial drug reinforcement. Notably, drug taking with initial reinforcement involves a potentiation of the projection from prefrontal cortex (PFC) to nucleus accumbens (NAc), while other glutamatergic projections are mostly involved in craving, like basolateral amygdala (BLA)-NAc projection, or in withdrawal/negative symptoms, like paraventricular thalamus (PVT)-NAc projection. With increasing administration of drugs of abuse and progressive shift toward compulsive abuse, the dorsal (dorso-lateral) striatum seems more and more implicated, with dopaminergic cells involved shifting progressively from the VTA to the substantia nigra pars compacta (SNc) [ 4 ]. Recently, data acquired through optogenetic dopamine neuron self-stimulation suggested prominent synaptic strengthening of the orbitofrontal cortex (OFC) to dorsal (dorso-medial) striatum projection in compulsive mice [ 31 ].

Drug addiction is conceptually defined as a three-stages cycle: (1) consumption/binge/intoxication, (2) withdrawal with its negative affect and (3) craving stage (Fig. 2 ) [ 27 ]. Animal models and human imaging studies have exposed the different brain areas involved in each of these stages. Briefly, the VTA-NAc (for reinforcement) and dorsal striatum (for stimulus-response habits) are important for the consumption/binge/intoxication stage, the extended amygdala with the hypothalamus and the brainstem in the withdrawal stage and cortical areas, the dorsal striatum, the hippocampus and the basolateral amygdala in the craving stage (Fig. 2 ).

figure 2

Progression to addiction is defined as a transition between three consecutive phases [ 252 ]: (1) Recreational, sporadic drug taking, in which drug of abuse administration is occasional and one activity among many other distractions of the individual. (2) Intensified and sustained drug use, in which drug administration strengthens and becomes the principal recreational activity of the individual; at this phase drug taking becomes a habit. (3) Loss of control of drug use and addiction, in which drug seeking and taking are now the principal activity of the patient. The first phase can occur to every person as drugs of abuse hijack the same brain circuit as natural rewards. The second phase occurs only in vulnerable users. The phase of addiction seems to be due to a second vulnerable trait with loss of control and compulsivity. Three stages of addiction are described [ 27 ]: (1) Binge/intoxication stage: reinforcing effects of drugs may initially use mainly dopamine and opioid peptides in the nucleus accumbens (NAc) and involves the ventral tegmental area (VTA). Subsequently, cue–response habits develop and includes the substantia nigra pars compacta (SNc) and the dorsal striatum. (2) Withdrawal/negative affect stage: the negative emotional state of withdrawal may involve the extended amygdala with corticotropin-releasing factor (CRF), norepinephrine and dynorphin as key neurotransmitters. Main projections of the extended amygdala consist of the hypothalamus and brainstem. (3) Craving stage: this stage includes conditioned reinforcement in the basolateral amygdala (BLA) and contextual processes in the hippocampus. This is controlled by cortical areas (prefrontal cortex (PFC) and orbitofrontal cortex (OFC)). A key neurotransmitter involved in the craving stage is glutamate.

The progression of drug addiction begins with the first exposure, mostly when the drug is taken voluntarily for its recreational and hedonic effect, and progressively consolidates during repeated but still controlled drug use. While administration intensifies along with loss of control over drug intake, drug use becomes habitual and compulsive in vulnerable individuals [ 4 , 29 , 30 ] (Fig. 2 ). This progression from voluntary drug intake to habitual and compulsive use represents a progression from ventromedial to more dorsolateral regions of the striatum and from prefrontal cortex (PFC) to orbitofrontal (OFC) and more global cortical region [ 4 , 31 ] (Fig. 1 ).

Synaptic plasticity

Brain plasticity is a fascinating capacity allowing appropriate modification of the neural activity in response to new experiences and environmental stimuli [ 32 ]. Modifying the synaptic strength between neurons is widely assumed to be the mechanism by which memory is encoded and stored in the brain [ 7 ]. Hence, it is appealing to hypothesise that drugs of abuse cause long-term alterations on behaviour by changing synaptic plasticity in key brain circuits [ 4 , 7 , 32 ].

Drugs of abuse such as cocaine induce specific synaptic plasticity in the mesolimbic circuitry. One single injection of an addictive drug can already modify the excitatory synaptic strengths in the VTA. Indeed, it has been extensively shown that the AMPA/NMDA ratio is increased in VTA dopamine neurons after one dose of cocaine and that some glutamate AMPA receptor 2 (GluA2)-containing AMPA receptors (AMPARs) are exchanged for GluA2-lacking ones [ 33 , 34 ]. At the same time, NMDA receptor (NMDAR) function decreases. All these elements cause an impairment in eliciting long-term potentiation (LTP). Different types of synaptic plasticity in VTA dopamine neurons induced by rewarding and aversive experiences are comprehensively reviewed by Pignatelli and Bonci [ 35 ]. Midbrain dopamine neurons are central in the mesolimbic circuitry for both natural rewards and drugs of abuse [ 18 , 36 ]. The VTA is known to be a central hub integrating numerous inhibitory inputs as GABAergic synapses represents 50–80% of all synapses onto VTA dopamine neurons [ 37 , 38 , 39 ]. GABAergic inhibition of dopamine neurons is mediated by both fast ionotropic GABA A receptors and slow metabotropic GABA B receptors [ 40 ].

In 2017, Edwards et al. [ 41 ] showed that the principal monosynaptic projection to VTA dopamine neurons arising from the NAc [ 42 ] inhibits the firing of dopamine neurons via activation of GABA B receptors, whereas local VTA inhibitory interneurons inhibits dopamine neurons through GABA A receptors. Today, it is well established that pharmacological activation of GABA B receptors (e.g. by baclofen) reduces cue-associated cocaine craving as well as reduce cocaine use in humans [ 43 , 44 , 45 ] and it reduces rewarding and reinforcing effects of cocaine on animal models [ 46 , 47 , 48 , 49 ]. Edwards et al. report [ 41 ] indicates that the therapeutic effects of baclofen might pass through VTA dopamine neurons’ GABA B receptors. Intrathecal Baclofen is an effective and safe long-term treatment used worldwide to treat severe spasticity [ 50 , 51 ]. Oral baclofen is less effective and has significant rates of side effects, like sedation, somnolence, vertigo and headache especially when prescribed off-label for drug addiction (because higher doses are commonly used) [ 51 , 52 ] . . Indeed, contrasting results on the effect of baclofen in reducing alcohol craving [ 53 , 54 , 55 ] and cocaine dependence [ 43 , 56 ] were probably due to different severity of alcohol dependence of the enroled patients. This is way higher dose are tested and often prescribed off-label for drug addiction [ 55 ]. Thus, self-poisoning that could lead to severe toxicity and death represents one the major concern of baclofen use in drug addiction. Therefore, baclofen should be prescribed with caution and close monitoring [ 52 , 57 ].

Together with drug of abuse-induced LTP at excitatory synapses, plasticity of GABAergic inhibitory synapse in the VTA also have an impact on the firing rate of VTA neurons, at least following opioid [ 58 ] and cocaine administration [ 59 ]. Normally, NMDA activation, during excitatory LTP (induced by high-frequency stimulation), leads to the release of NO that will activate guanylate cyclase in adjacent GABAergic terminals, which in turn, leads to increase in GABA release. This presynaptic NMDA receptor-dependent GABAergic LTP heterosynaptic plasticity, is named LTP GABA . Nugent el al. [ 58 ] showed that opioids blocks LTP GABA through a disruption of the coupling between nitric oxide (NO) and guanylate cyclase. The incapability of GABAergic synapses to potentiate after morphine or cocaine administration may promote LTP of glutamatergic synapses [ 58 , 59 ]. The early loss of inhibitory control combined with potentiation of glutamatergic synapses on dopaminergic neurons might represent adaptations that increase vulnerability to addiction [ 58 , 59 ]. Furthermore, GABA A receptor modulators modify the addictive drugs effects [ 60 , 61 ], and targeting these receptors might be seen as an effective therapeutic strategy but precluded by many side effects among which dependence itself [ 62 , 63 , 64 ].

In addition to the discovery of LTP GABA , Nugent’s group showed that morphine is also able to modulate a form of postsynaptic LTD (LTD GABA ) at GABAergic synapses onto VTA dopamine neurons. Remarkably, after a single administration of morphine, LTD GABA was absent in slices from morphine-treated rats while unaffected in slices from saline-treated rats, indicating a bidirectional control of morphine on GABAergic synaptic plasticity in the VTA [ 65 ]. This absence of LTD GABA is suggested to result from an occlusion effect due to prior morphine-induced decrease in GABAergic synaptic strength through potentiation of glutamatergic transmission and mediated by endocannabinoid signalling [ 66 ]. It is also possible that morphine alters the ability of synapses to exhibit evoked LTP or LTD in the VTA. Previous experiences such as exposure to drugs of abuse, stress, visual or sensory deprivation can change the ability of synapses to undergo subsequent plasticity in response to LTP and LTD induction protocols. This concept of modification of plasticity capability is referred as metaplasticity [ 67 ].

In the NAc, chronic exposure to addictive drugs induces specific synaptic changes that are different from those of the VTA, including a decrease of the AMPA/NMDA ratio as some AMPARs are endocytosed. This leads to a depressed synapse (sometimes referred as long-term depression (LTD) like state), where NMDAR-dependent LTD is reduced or, in some experiments, abolished [ 68 , 69 ]. Highlighting the importance of temporal aspects, studies of withdrawal period after chronic administration of cocaine, showed that synaptic AMPAR levels increase during the first week of withdrawal and persist elevated for weeks [ 70 , 71 , 72 ]. It is established that cocaine challenge transiently decreases AMPAR surface expression, while AMPARs recover back to upregulated levels within a week, with a continuous increase during what is known to be the incubation of craving stage [ 73 ].

The abstinence period after withdrawal is of particular interest considering the classical progression of the disease, the chance of relapse and the opportunity for new therapeutic targets. A seemingly counterintuitive concept named ‘incubation of cocaine craving’ was introduced by Grimm et al. [ 74 ] who modelled cocaine-craving behaviour by using rats trained to press a lever to receive an injection of cocaine and were then forced in a withdrawal period where cocaine reward was no longer given. This concept of ‘incubation’ did not originate in drug addiction research but came from a four-stage model of the creative process proposed by Graham Wallas in 1926 [ 75 ]. Consistent with clinical observations in humans [ 76 , 77 , 78 ], they showed that relapse was progressively stronger over 2 months of cocaine withdrawal and suggest that a craving syndrome progresses or ‘incubates’ during the first 2 months of cocaine abstinence, and probably lasts for longer [ 74 ]. Subsequently, it was shown that this increase was due to the addition of new AMPARs lacking GluA2 and that these new receptors mediate the ‘incubation of cocaine craving’ [ 72 ]. Conrad et al. [ 72 ] showed that after extended withdrawal from cocaine, addition of synaptic AMPARs together with the increased conductance of GluA2-lacking AMPARs triggers higher sensitivity of NAc neurons to cocaine-related cues, leading to a strengthening of drug craving syndrome and relapse. In line with these results, it was suggested that as soon as abstinence is reached, the risk of relapse might be reduced if GluA2-lacking AMPARs were inactivated or removed from NAc synapses. It was thus proposed that GluA2-lacking AMPARs could be a new target for drug development for the treatment of cocaine addiction. While these calcium permeable AMPARs are also critical for the pathogenesis of numerous other neurological disorders (including epilepsy [ 79 ], fragile X syndrome [ 80 ], amyotrophic lateral sclerosis [ 81 ], Parkinson’s [ 82 ] and Alzheimer’s [ 83 ] diseases), developing drugs that specifically target them and not calcium-impermeable AMPARs, which are critical for normal CNS function, is challenging [ 84 ] (Fig. 3 ).

figure 3

DNA is packaged inside nuclei with the help of histones. These are positively charged proteins that strongly adhere to negatively charged DNA and form complexes called nucleosomes. Each nucleosome is composed of DNA wound around histone octomers (H2A, H2B, H3 and H4). Nucleosomes fold up to form chromatin fibre, which forms loops compressed and folded to produce fibres, which are coiled into the chromatid of a chromosome. Only by loosening compacted chromatin, the DNA of a specific gene can be made accessible to transcription. Some of these drug-induced modifications at the chromatin level are extremely stable and sustain the drug of abuse-induced long-term behaviours. Among them, histone post-translational modifications (PTMs) are known to be causally involved in drug-induced behaviours [ 194 ]. PTMs include acetylation (Ac), methylation (Me), phosphorylation (P), ADP ribosylation (PolyADP-R) and dopaminylation (DA), among a growing list of newly discovered modifications [ 162 , 172 ]. For example, while ubiquitylation (Ub) of H2A is known to be a key interactor of H3 methylation [ 253 ], its supposed role in drug addiction is still unknown. At this epigenetic level, some drugs were demonstrated to have an influence on drug-induces behaviours such as histone deacetylase (HDAC), bromodomain and DNA methyltransferase inhibitors. Locus-specific epigenome editing is now encouraging as a new field of investigation as it might help to the discovery of new specific and causal drug of abuse targets. Overview of the tetrapartite glutamatergic synapse composed of a medium spiny neuron (MSN), a glutamatergic projection, a glial cell and the extracellular matrix (ECM). Here, we focused on synaptic potentiation after drug of abuse administration with the addition at the post-synaptic membrane of glutamate AMPA receptor 2 (GluA2) lacking AMPA receptors (AMPARs). This mechanism might be reduced by metabotropic glutamate receptor 1 (mGluR1) positive allosteric modulator or more directly by GluA2-lacking AMPARs antagonists. In the same way, it was also shown that presynaptic mGluR2 agonists can potentially abolish drug seeking and impair craving incubation. Optogenetically-inspired 12 Hz deep brain stimulation (DBS) in the nucleus accumbens can also be a promising novel therapeutic for addiction. Finally, ceftriaxone, N-acetylcysteine, and inhibitor of matrix metalloproteases 9 (MMP-9), mainly through their action on glial cell and the ECM, are very interesting molecules that may be added in the addiction therapeutic arsenal.

Inspired by previous work performed in the VTA showing that metabotropic glutamate receptor 1 (mGluR1) LTD induces removal of GluA2-lacking AMPARs from synapses [ 33 , 34 ], Loweth et al. [ 85 ] demonstrated that synaptic GluA2-lacking AMPAR decrease could be accomplished by in vivo evoked mGluR1 LTD in the NAc. More importantly, their group showed that after prolonged cocaine or methamphetamine withdrawal, systemic injection of a mGluR1 positive allosteric modulator attenuated the expression of incubated craving by reducing GluA2-lacking AMPARs in the NAc [ 85 , 86 ]. These results suggest a strategy in which abstinent methamphetamine or cocaine users could use a systemically active compound to protect themselves against cue-induced relapse.

These latter studies were conducted without differentiating between D1 receptor D1R MSNs and D2R MSNs. In 2014, Pascoli et al. [ 87 ] demonstrated that this increase in the strength of excitatory afferents was exclusively related to D1R MSNs. Interestingly, the type of drug-evoked plasticity involved is also dependent on the input. It has been shown that even in the same D1R MSN a synapse connecting the PFC to the NAc increases its strength by inserting GluA2-lacking AMPARs whereas a synapse connecting the ventral hippocampus to the NAc increases the AMPA/NMDA ratio by inserting GluA2-containing AMPARs [ 87 ].

Besides operant self-administration, all these long-term synaptic modifications also underlie behavioural changes associated with drugs of abuse, such as locomotor sensitisation [ 88 , 89 ]. Locomotor sensitisation is a behavioural protocol used to model drug-induced behaviour [ 90 , 91 ]. In rodents, repeated cocaine injection induces gradually increased locomotor activity; after 5 days of consecutive injections, the locomotor response reaches a ceiling level. This state lasts for months after cocaine withdrawal [ 91 ]. As an experimental model, locomotor sensitisation is linked with increased tendency to self-administer psychostimulants [ 92 , 93 ] and with reinstatement of previously extinguished self-administration [ 94 , 95 ]. Whereas the existence of psychomotor sensitisation in humans is discussed [ 96 , 97 ], it is a key aspect of vulnerability to drug addiction and relapse, specifically drug craving or compulsive drug-seeking behaviour [ 91 , 98 , 99 ]. Still, locomotor sensitisation can be dissociated from the rewarding effect of a drug of abuse and conditioned place preference or self-administration are more appropriate experimental paradigms to test this aspect [ 100 , 101 , 102 ]. Even if drug-induced locomotor sensitisation is unclearly present in humans, as an animal model it offers a clear readout to understand the mechanisms by which drugs of abuse induce long-term brain modifications [ 91 ].

Furthermore, it has been elegantly demonstrated that optogenetic stimulation of the excitatory projections to the NAc is able to reverse cocaine and alcohol-evoked plasticity [ 87 , 88 , 89 ]. Briefly, applying a NMDAR or mGluR1-dependent LTD on cortico-accumbal glutamatergic synapses, before a drug of abuse administration, diminishes its effect. In another study, Luscher’s team took advantage of the knowledge, obtained from optogenetic in vivo experiment in rodents, to implement a novel deep brain stimulation (DBS) protocol that abolishes behavioural sensitization to cocaine (and thus that would be efficient during the relapse phase) [ 103 ]. Basically, the idea is to manipulate synaptic plasticity in the NAc to reverse pathological synaptic transmission and its associated behaviours following exposure to drugs of abuse. In this study, as a therapeutic use of optogenetic tools in humans is for now inapplicable [ 104 ], the authors reversed cocaine-evoked plasticity and thus drug-induced behaviours by using DBS instead of optogenetic. Indeed, DBS is routinely used in clinic and a new DBS protocol can easily be translationally implemented to the human therapeutics [ 105 , 106 ]. They refined the classical high-frequency DBS protocol (that has no sustained effect on cocaine sensitization, probably because it does not affect synaptic plasticity) by applying a low frequency stimulation (12 Hz to equal the one used in the optogenetic endocannabinoid- dependent LTD protocol) in the NAc together with the administration of a D1R antagonist necessary to unmask the mGluR-dependent LTD in D1R MSNs as demonstrated previously [ 107 ] (Fig. 3 , see section on clinical treatment for broader discussion on DBS).

Kalivas’ group showed in 2009 [ 108 ] that after extended withdrawal from chronic cocaine self-administration, cocaine-induced metaplasticity at the excitatory synapses in the NAc that impairs the ability of PFC stimulation to produce LTP or LTD in NAc MSNs. They also showed that N-acetylcysteine reverses cocaine-induced metaplasticity, allowing the induction of both LTP and LTD and that N-acetylcysteine decreases cocaine-relapse in a rodent model. We are currently awaiting the results of a randomised and control study that is testing newly detoxified (and therefore abstinent) hospitalised patients who received a 3–4 week course of treatment, in order determine if N-acetylcysteine can be a useful medication candidate to avoid relapse in patients with cocaine dependence (NCT03423667).

GABAergic D1R and D2R MSNs, equally compose and are mosaically intermingled throughout the striatum [ 109 ]. As explained above, D1R and D2R MSNs send axonal projections outside the striatum, forming the two main output pathways, respectively the direct and indirect pathways [ 16 , 17 ]. In a certainly oversimplified model, the activation of the D1R MSNs result in facilitation of locomotion, reward, and reinforcement while the activation of D2R MSNs result in opposing effects [ 110 , 111 , 112 , 113 ]. In addition to the long-range projections, these neurons form short-range synaptic connections with one another within the striatum, and because they consist of inhibitory collaterals, a mechanism known as lateral inhibition [ 114 , 115 , 116 , 117 ]. Interestingly, these connections are not symmetrical, with D2R MSNs forming more synaptic connections on D1R MSNs [ 115 , 117 ]. Through this previously understudied collateral transmission, Dobbs et al. [ 115 ] presented a novel mechanism by which cocaine exerts its stimulant effect: cocaine, by blocking DAT receptors enhance levels of dopamine and subsequently activating D2Rs, causes a suppression of lateral inhibition and thus disinhibition of D1R MSNs in the NAc which in turn promotes locomotion [ 115 ]. Furthermore, Alvarez’ group suggested that constitutive low D2R levels, through imbalanced lateral inhibition, might pre-sensitised D1R MSNs, facilitate behavioural plasticity to repeated cocaine and promotes an addiction vulnerable phenotype [ 116 ].

The characterisation of the role of glia and the extracellular matrix (ECM) in drug-induced synaptic plasticity is an exciting emerging field of drug addiction research as it comes with promising new therapeutic possibilitiess [ 118 , 119 , 120 ]. Mulholland et al. [ 118 ] summarised and emphasised the role of the ECM and of astroglial cells in the regulation of synaptic plasticity. Of great interest, restoring downregulated glutamate transporter 1 (EAAT2) with ceftriaxone reduces drug seeking in animal models [ 121 , 122 ]. Matrix metalloproteases (MMP) are important regulators of the ECM and contribute to synaptic plasticity [ 123 ]. Inhibiting their activity result in suppression of the reinstatement of cocaine conditioned place preference [ 124 ] and selectively inhibiting MMP-9 prevents cue- and cocaine-induced reinstatement of cocaine self-administration [ 119 ]; these results open additional therapeutic possibilities with the use of inhibitors of MMP-9 as an innovative targeted approach [ 119 , 124 , 125 ] (Fig. 3 ). Still, at our knowledge, there are no randomised controlled study currently investigating these ECM-related drugs.

Drugs of abuse-induced modifications in glutamatergic nuclei targeting the NAc, or the VTA and essential part of the reward circuit, are less studied than cortico-striatal synapses despite the fact that they play a crucial role in the development of drug addiction. Indeed, in the OFC and PFC, chronic alcohol exposure significantly increases LTP in pyramidal neurons [ 126 , 127 ]. Kazanetz et al. [ 128 ] showed that repeated cocaine injections impair endocannabinoid-LTD and mGluR2/3-LTD in the PFC. They postulated that this might mechanistically participate in the induction of a postsynaptic, observed LTP-like phenomenon with an enhanced AMPA/NMDA ratio. It was also demonstrated that neurons of the infralimbic cortex present a decrease in mGluR2 [ 129 ]. In addition, alcohol-dependent rats exhibit an escalation of ethanol seeking, which was abolished by restoring mGluR2 expression in the infralimbic cortex via viral-mediated gene transfer [ 129 ]. Notably, mGluR2 agonist was shown to impair the incubation of cocaine craving [ 130 ] and to attenuate reinstatement of cocaine-seeking [ 131 , 132 ](Fig. 3 ). Recently, Caprioli et al. [ 133 ] extensively reviewed preclinical studies on allosteric modulators of mGluRs on animal models of drug addiction and their potential translational implications. The results reviewed [ 133 ] indicate an remarkable effect of allosteric modulators of presynaptic mGluR2 and possibly mGluR7, supporting the idea that these compounds should be tested as potential medications for addiction treatments.

Besides the PFC, other brain regions appear to be key areas in drug addiction as the paraventricular thalamus (PVT) - a central hub for cortical, sensory and limbic information [ 134 , 135 , 136 , 137 , 138 , 139 , 140 ]. In 2016, Zhu et al. [ 141 ] showed that chronic morphine administration potentiates excitatory synapses between the PVT and D2R MSNs via insertion of GluA2-lacking AMPARs. Remarkably, in vivo optogenetic depotentiation at these synapses abolishes morphine withdrawal symptoms. In a recent paper, projections from the PVT to the NAc were shown to be critical for augmentation of heroin seeking in food-restricted rats [ 142 ] (Fig. 1 ). Actually, Otis et al. [ 143 ] demonstrated that the PVT is an integrative hub for reward seeking behaviour and that PVT-NAc neurons integrate different inputs from the PFC and the lateral hypothalamus to precisely guide reward seeking behaviour. In a recent review, De Groote et al. [ 140 ] focused on the new advances in the understanding of the roles of the PVT-NAc connections in motivated behaviours, highlighting their implications in drug addiction.

Drug addiction-related genes and transcriptomic regulation

Modifications in gene expression contribute to the long-lasting effect sustaining drug addiction; thanks to gene-expression arrays, RNA-sequencing and candidate gene approaches, the specific genes and their regulatory transcriptomic mechanisms involved in drug addiction development and maintenance are now better understood.

Drug addiction-related genes

For example, the use of conditional gene knockout in mice emphasises the importance of monoamine membrane transporters (dopamine transporter, and serotonin transporter) [ 144 , 145 ] and of mGluRs [ 146 , 147 ]. As new animal models of drug addiction, these approaches are also useful to better characterise fine-tuning of important pathways involved in addiction. For example, a scaffold protein known as Maged1 has been shown to be involved in cocaine reward and reinforcement [ 148 ]. We demonstrated that Maged1 inactivation impairs drug-evoked dopamine release and glutamatergic synaptic plasticity in the NAc. Inactivation of Maged1 in mice was able to abolish behavioural sensitization to cocaine as well as cocaine conditioned place preference and operant self-administration behaviours [ 148 ]. This sole genetic alteration, causally linked to a strong alteration of drug-induced behaviours, impairs (at least) two core neuronal mechanisms leading to addictive behaviours: (1) cocaine-evoked release of dopamine in the NAc and (2) NAc plasticity, with a reduced AMPA/NMDA ratio and a resistance to LTD. Actually, it seems that, after Maged1 inactivation, the excitatory synapses in the NAc shift to a depressed state. Our hypothesis is that, in line with the previously discussed in vivo optogenetic induced LTD, this impairment could be a key factor for the significant decrease in sensitization to psychostimulants [ 87 , 103 , 148 ]. Actually, it seems that placing neurons in a state of ‘presensitization’ is able to prevent drug-induced sensitization itself [ 148 , 149 ]. Our group is now trying to understand what are the cellular and molecular pathways directly altered by Maged1 inactivation and responsible for this strong anti-addictive drug phenotype. Remarkably, the promoter of Maged1 was found in a list of 213 promoters that co-precipitate with acetylated histones and with the activated form of cAMP response element binding protein (CREB) after chronic drug taking [ 150 ]. In line with this result, preliminary and unpublished results from our laboratory point out a specific epigenetic mechanism, in parallel with an alteration of synaptic plasticity in excitatory projection to the NAc, that would link Maged1 to its major effect on drug-induced behaviours. This selected gene approach is of great interest in refining our knowledge of pathways hijacked by addictive drugs. Using cell sorting of D1R MSNs and D2R MSNs as described previously [ 151 ], our group also identified the G-protein-regulated inducer of neurite outgrowth 3 (GPRIN3) in both MSN populations but strikingly more expressed in D2R MSNs [ 149 ]. The GPRIN family (GPRIN1, GPRIN2 and GPRIN3) are Gαi/o-regulated proteins suggested to intermediate the communication between GPCRs and the sequential intracellular target [ 152 ]. Indeed, GPRIN1 and GPRIN2 have been described as alternative (to adenylyl cyclase) mediators of GPCRs signalling but GPRIN3 had a much less defined role [ 152 , 153 ]. To understand the role of GPRIN3 in the pathophysiology of the D2R-indirect pathway, we induced a D2R-MSNs-specific knockdown (KD) of GPRIN3 using small hairpin RNA and lentiviruses [ 151 , 154 ]. We first observed a significant increase in distal branching, the points of convergence between glutamate and dopamine synapses in MSNs [ 155 ] and also key targets of cocaine, which itself promotes increase in distal branching in the NAc of mice [ 156 , 157 , 158 , 159 ]. Thus, we tested the cocaine acute effect and locomotor sensitization and observed a decrease in cocaine-induced hyperlocomotion after inactivation of GPRIN3 using a CRISP/Cas9 approach. The significant increase in distal branching in GPRIN3 D2R-MSNs KD corroborates our hypothesis that the lack of GPRIN3 induces a ‘presensitization process’, able to change the targets of cocaine and therefore altering its effects [ 149 ]. Finally, we provide the first evidence that GPRIN3 partners with D2R in the striatum and modulates cocaine-induced behaviours [ 149 ].

Transcriptomic and epigenetic regulations

Epigenetics is a broad field and has multiple definitions that comprise several biochemical mechanisms (including DNA methylation and histone modifications) sustaining modifications in gene expression throughout the lifecycle of an organism without mutations of the DNA itself [ 160 , 161 , 162 ]. Epigenetics can be considered as the process through which environment (and normal development) interacts with an individual’s genome to determine all phenotypic traits, in health and disease. Stable modifications in gene expression are also said to be ‘epigenetic’, because they are heritable in the short term (through mitosis) [ 160 ] and in some cases trans-generationally, thus, providing a potential mechanism for environmental influences to be passed from parents to offspring [ 163 , 164 , 165 ]. Handel and Romagopalan [ 163 ] mentioned that “epigenetics allows the peaceful co-existence of Darwinian and Lamarckian evolution”. Such trans-generational epigenetic inheritance of drug addiction vulnerability remains debatable [ 161 ], but has been increasingly studied for the last 20 years [ 166 , 167 ]. Some epigenetic changes are very stable, an thus mediate both drug addiction susceptibility and drug-induced brain alterations that underlie the development of drug addiction [ 161 ].

As the NAc is seen as the central hub of drug addiction, with the notion that chronic drug use induces long-lasting structural, electrophysiological and transcriptional changes in the NAc, researchers are mostly targeting epigenetic modifications in NAc cells. Still, considering initial reports of cocaine-induced epigenetic modifications [ 168 , 169 ], it might be relevant to study further epigenetic changes in other regions such as glutamatergic inputs to the NAc, and further in the VTA, as they are implicated in the physiopathology of drug addiction [ 170 , 171 ] as mentioned above.

To date, the three main epigenetic mechanisms consist of (1) DNA methylation, (2) action of the non-coding RNAs and (3) histone post-translational modifications (PTMs). As an illustrative example, we will focus here only on histone PTMs. PTMs of histone residues on their N-terminal tails, that protrude from the nucleosome core, control chromatin condensation and the switch between euchromatin and heterochromatin and thus DNA-accessibility and gene expressions. PTMs include acetylation, methylation, phosphorylation, ADP ribosylation, ubiquitylation and sumoylation, among a growing list of newly discovered modifications [ 162 , 172 ].

Among these PTMs, the most studied is the acetylation of H3 and H4, that is increased in the NAc after chronic exposure to drugs of abuse [ 150 , 173 , 174 ]. This increase in global acetylation levels is the result of drug-induced alterations in the balance of histone acetyltransferase and histone deacetylase (HDAC) function and is associated with gene activation. CREB-binding protein, a histone acetyltransferase critical to memory processes [ 175 ], is required for cocaine-induced increases in histone acetylation in the NAc [ 176 ].

Fifteen years ago, Tsankova et al. [ 177 ] showed that imipramine, a monoamine reuptake inhibitor used for decades to treat depression, was effective through histone remodelling in depression and highlight the therapeutic potential for chromatin regulation with histone methylation and deacetylation inhibitors in depression. Nevertheless, like with synaptic plasticity (see above), discovering a drug that would interfere with epigenetic mechanisms and thus decrease drugs of abuse effect faces temporal aspects issues [ 173 , 176 , 178 , 179 , 180 , 181 , 182 ]. Indeed, timing has a strong impact considering conflicting results obtained after experimental manipulations of histone acetylation. An acute administration of HDAC inhibitors systemically or directly into the NAc, promotes behavioural responses to the drugs. However, prolonged administration decreases cocaine behavioural effects. In 2013, adding a new layer of complexity, Kennedy et al. provided comprehension to this time-dependent regulation [ 183 ]. Remarkably, they showed that prolonged intraNAc administration (but not acute administration) of a HDAC inhibitor attenuated cocaine behavioural effects by inducing a form of repressive histone methylation. This study showed, for the first time, cross-talk among different types of histone modifications [ 183 ]. Besides cross-talk between different epigenetic modifications, multiple modifications work in parallel and there is often a decoupling between an observed modification at a specific locus and its final transcription [ 161 ]. Decoding these chromatin marks will be a future challenging field. Like with HDAC inhibitors, there are promising findings based on the use of DNA methyltransferases inhibitor [ 184 , 185 ] (Fig. 3 ). Though, the main issue with these new potential treatments for drug addiction is their lack of specificity. One of the key challenge for the pharmaceutical industry will be to generate small molecules with more specific targets [ 6 ].

While histone acetylation and methylation are increasingly studied, an important field of future investigation will be to understand the other drug-induced histone PTMs. It already seems that chronic cocaine alters levels of histone phosphorylation [ 174 , 186 , 187 ], and poly-ADP ribosylation [ 188 ]. Recently, an unexpected role for the intracellular dopamine in VTA has been revealed, showing that DA interacts with chromatin to initiate a new form of epigenetic regulation called dopaminylation [ 189 ] (see Table 1 for a summary of cocaine-related epigenetic modifications).

Further studies showed that histone PTMs that occur in the NAc after chronic drug administration are locus specific [ 150 , 190 , 191 ]. Even though, drugs of abuse alter global levels of multiple histone PTMs, such as increased histone acetylation or decreased methylation in the NAc, genome-wide studies have confirmed that a greater number of genomic sites show increased acetylation [ 150 ] or decreased methylation [ 190 , 191 ]. Conversely, hundreds of genes show opposite or no changes in these same PTMs after drug exposure. What defines whether, and in which direction, a specific gene is modified in the context of a global histone PTM is an intriguing and unsolved question [ 161 ]. These genome-wide studies (ChIp on chip or ChIpSeq) are nowadays fundamental to understand where PTMs and other epigenetic modifications are deposited. This will be fundamental to guide new therapeutics.

Actually, with new tools such as zinc finger proteins (ZFPs) DNA-binding domains and, more recently, RNA-guided CRISPR/dCas9 (drastically easier to design) [ 192 , 193 ], it is now possible to control epigenetic modifications at a single gene in a specific type of cell in a specific brain region [ 162 ]. Heller et al. demonstrated that gene-targeted epigenetic editing (targeted to the Fosb [ 194 ] and Cdk5 [ 195 ] locus with ZFP technology) can alter drug-related behaviours [ 194 , 195 ]. This represents crucial evidence that gene-specific changes to the epigenome are not simply correlated, but rather causal, in regulating transcriptional responses to drugs of abuse administrations. These new results of “causal epigenomics” are very encouraging as they open the way to precise translational therapeutic approaches for drug addiction and other CNS diseases.

Linking epigenetics and synaptic plasticity

Today, most studies investigate synaptic plasticity and epigenetic as two distinct fields and it is not clear how these research topics are connected to each other. Understanding how epigenetics is connected to synaptic plasticity is an emerging research issue [ 6 ].

Of course, bridging epigenetic mechanisms with synaptic plasticity is not limited to drug addiction field. For example, in 2011, Monsey et al. [ 196 ] elegantly demonstrated that DNA methylation and histone H3 acetylation regulate auditory fear conditioning and its related synaptic plasticity in the amygdala. In 2014, Massart et al. [ 197 ] suggested that sleep deprivation induces epigenetic modification (alteration in DNA methylation and hydroxymethylation) that triggers synaptic plasticity modifications by changing expression of plasticity related genes.

Regarding drug addiction, some epigenetic marks seem fundamental and upstream as illustrated by HDAC inhibitors effect on drug-induced synaptic and behavioural modifications [ 178 , 198 , 199 , 200 ]. Additionally, Maze et al. [ 201 ] demonstrated morphological plasticity induced by cocaine through the histone methyltransferase G9a. Again advocating for causal epigenetic, Authement et al. [ 66 ] demonstrated that HDAC inhibition locally in the VTA is sufficient to reverse epigenetic modifications and synaptic plasticity changes after morphine administration.

Two transcription factors implicated in addiction exemplify this bridging attempt: CREB and ∆FosB (a truncated form of the FosB gene) are both activated by several drugs of abuse [ 202 ]. CREB activation occurs in both subtypes of NAc MSNs (D1R and D2R), while ∆FosB activation is limited to D1R MSNs in response to all drugs of abuse except for opioids, which remarkably induce the protein in both MSNs [ 203 ]. Expression of active CREB in NAc MSNs increases their excitability [ 204 ] and underlies drug-induced long-term synaptic plasticity and associated changes in dendritic spine plasticity [ 205 ]. ∆FosB is also linked to synaptic plasticity but evokes contrasting effects on the two MSN subtypes, with increased AMPA receptor function induced in D1R MSNs and decreased AMPA receptor function induced in D2R MSNs [ 206 ]. Renthal et al. [ 150 ] unravelled CREB and ∆FosB target genes and observed that these genes are mainly involved in neuronal excitability and synaptic function. Moreover, as already briefly discussed above, CREB and ∆FosB action have also been related to multiple epigenetic regulations, including histone acetylation and methylation [ 150 ]. Besides, a novel mechanism for bridging the gap between epigenetic control of transcription and synapse plasticity might be seen in microRNAs [ 207 ]. The most studied miRNA in the context of synaptic plasticity is miR-132 and is known to be CREB-dependent [ 208 ]. In the striatum, miR-212 targets the epigenetic regulator methyl CpG binding protein 2 (MeCP2). MeCP2 acts as a transcriptional repressor through recruitment of histone deacetylases to methylated DNA segments [ 209 , 210 ].

Clinical treatments for drug addiction

Besides psychosocial interventions [ 211 ] such as cognitive behavioural therapy, the most widely used treatment for drug addiction involves agonist-like medication, a solution inadequately called replacement or substitution therapy [ 212 ]. This type of treatment has been successfully implemented in the daily practice for opioid use disorder (e.g.: methadone, buprenorphine) [ 213 ] and tobacco use disorder (e.g.: nicotine patch or gum, varenicline) [ 214 ]. Currently, this agonist-like treatment is also promising for psychostimulant use disorder [ 215 ]. Still, considering the addictive drug-like effect, the risk of abuse, misuse and diversion, replacement therapy should be prescribed with caution [ 215 , 216 ].

Recently, a randomised and control study on a cocaine vaccine failed to show an effectiveness but instead raised an important issue: immunised subjects may have increased their cocaine use to overcome the competitive anti-cocaine antibody inhibition [ 217 ]. Even though significant improvements have been developed for immunopharmacotherapies for psychostimulant addiction over the last decade, very few candidates have been evaluated so far in clinical trials [ 218 ]. These considerations are some of the reasons why other treatments for drug addiction should emerge with the help of neurobiological research [ 219 ].

Following successful subthalamic nucleus DBS for Parkinson’s disease [ 220 , 221 , 222 ], DBS was investigated for diverse psychiatric diseases including depression [ 223 ], obsessive-compulsive disorder [ 224 ] and Tourette syndrome [ 225 ]. Today, indications for DBS are enlarging, with several positive case reports and small cases series that studied NAc DBS for drug addiction. The first studies showing potential positive effects on drug addiction were reports on application of NAc DBS primarily intended for other medication-refractory neuropsychiatric disorder where a comorbid drug addiction was unexpectedly resolved [ 226 , 227 ]. For DBS treatment in drug addiction, it seems that clinical empirical results led to further bench investigations and refinement [ 88 , 103 , 228 , 229 , 230 , 231 ], or at least, clinical and animal studies evolved in parallel with poor connectivity between the two.

Afterwards, many case reports and small cases series studied NAc DBS being used primarily for drug addiction, all showing encouraging decreases in drug use [ 232 , 233 , 234 , 235 , 236 , 237 ]. However, these studies are limited by their descriptive nature, inconstant follow-up, multiple publication bias, small patient numbers and lack of blinded stimulation and standardised outcome measures. At this stage, additional preclinical and clinical research are needed to clarify the role of DBS in the treatment of drug addiction [ 237 ]. Currently, randomised and control clinical studies are conducted (NCT01245075).

In a recent review, Sanna et al. [ 238 ] highlighted how repetitive transcranial magnetic stimulation (rTMS) confirms the hypodopaminergic hypothesis of drug addiction. While enhancing dopaminergic function through direct or indirect pharmacological approaches does not significantly alleviate symptoms, in numerous studies, and has not yielded a single FDA-approved medication [ 239 ], rTMS might indirectly modulate the dopaminergic system. Many rTMS studies stimulate the dorsolateral PFC [ 240 , 241 ] that projects to the VTA and thus induces an increase in dopamine release in the synaptic cleft in the NAc [ 55 , 242 , 243 ]. Nevertheless, considering the heterogeneity of methods used in rTMS studies during the last 10 years [ 238 ], protocols and guidelines, were recently suggested by an international network of experts in neuromodulation and addiction to improve homogeneity of studies [ 244 ]. From this report, it is clear that multiple technical details for optimal stimulation need further investigations that might be achieved through preclinical studies. For example, low frequency (but not high-frequency) rTMS before methamphetamine exposure in rats blocked drug-induced conditioned place preference [ 245 ]. Being non-invasive, with insignificant side effects, rTMS could be seen as a great opportunity for drug addiction treatment. We are currently waiting for the results of a randomised and control study that aims at determine if, in heavy alcohol users, a single session of TMS can lower a patient’s craving and brain response to alcohol cues (NCT02939313).

Interesting views of clinical treatments for drug addiction are discussed in some other reviews [ 212 , 215 , 216 , 219 ]. The clinical impact of new treatments also depends on their translation into clinical practice which is mainly promoted by the pharmaceutical industry [ 219 ]. Indeed, even when an effective treatment is identified through basic research, it is commonly challenging to translate it to clinical practice, as illustrated by naltrexone as a treatment of alcoholism [ 219 ]. Another example of problematic translation to clinic is illustrated by modafinil, a treatment that has been reported to attenuate cocaine euphoria but for which larger clinical randomised and controlled studies showed controversial results [ 246 , 247 ].

Future directions

Drug addiction is a brain disease strongly influenced by environment and psychosocial aspects. The psychosocial conditions in which it has developed are extremely important. Exposure to conditioned cues can be a central issue in causing drug cravings and relapses, even after successful treatment, and thus they have to be minimised [ 2 , 74 , 77 ]. The pathophysiological aspects are particularly unsteady. For instance, as discussed in this review and in other ones [ 73 , 248 ], synaptic plasticity is dynamically altered after psychostimulant administration, so that a treatment could have opposite effects depending on timing aspects of the administration protocol. In addition, a prolonged treatment may involve compensatory mechanisms, giving unexpected results (e.g.: when HDAC inhibitors and psychostimulants are both administered acutely, they have synergistic effects through hyperacetylation and thus transcriptional activation of psychostimulant-regulated target genes. Conversely, when a drug of abuse is given in the context of chronic HDAC inhibitor, compensatory mechanisms may promote acetylated histone to the promoters of genes responsible for inducing histone methylation and thus chromatin condensation and gene repression, all of which, in turn, gave opposite effect [ 183 ]). Thus, the evolution through the different stages of the disease has to be taken into account [ 249 ] and treatment must follow them. These two aspects have to be incorporated in a holistic treatment strategy. Besides, studying combination of different cutting-edge approaches, with animal models of addiction, such as targeted rTMS or DBS with more systemic epigenetic modulation might show a better restoration of altered synaptic transmission and decrease the probability of relapse in drug addiction. Basically, drug addiction is a disease that seems to be difficult to treat preventively but it is more conceivable to help patients that would be in an abstinence stage not to experience relapse of their disease. As addiction is chronic and relapsing, a good treatment outcome is a significant reduction of drug administration and long periods of withdrawal, with only sporadic relapses [ 2 ].

It is clear that the main issues for optimal therapeutic management of this specific psychiatric disease belong to its dynamic complexity, diverse temporal evolution and undeniably psychosocial aspects. In this review, we focused mostly on the effects of drugs of abuse on synaptic plasticity and epigenetic modifications. Nowadays, these two subfields are mostly studied separately and the understanding of how these two main addictive drug-induced brain modifications interact might be fundamental for addiction research [ 6 ]. Indeed, argument for clinical trials for new treatments emerge from fundamental behavioural studies that should be implemented in a global approach to the addicted patient.

Conclusions

Here, we highlight, from a vast fundamental literature (mainly based on rodent models), promising therapeutics that would potentially treat drug addiction. Based on effect, on synaptic plasticity and epigenetic mechanisms, treatments such as GluA2-lacking AMPAR antagonists [ 72 , 84 ], mGluR1 positive allosteric modulator [ 85 ], NAc 12Hz-DBS [ 103 ] (in line with other promising neuromodulation therapeutics such as rTMS or transcranial direct current stimulation [ 250 ]), N-acetylcysteine [ 108 ], HDAC inhibitors [ 183 ] or even (in very early stages of investigation) CRISPR/dCas9 epigenetic editing [ 194 , 195 ] could be potential candidates for human randomised clinical trials (Fig. 3 ).

Finally, it is fundamental to consider the specific clinical aspects of the disease that would help to develop a personalised-treatment strategy. Indeed, after going from the bench to the bedside it will also be essential to assess the reversed route.

Roth GA, Abate D, Abate KH, Abay SM, Abbafati C, Abbasi N, et al. Global, regional, and national age-sex-specific mortality for 282 causes of death in 195 countries and territories, 1980–2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet. 2018;392:1736–88.

Article   Google Scholar  

Leshner AI. Addiction is a brain disease, and it matters. Science. 1997;278:45–47.

Article   CAS   PubMed   Google Scholar  

Nielsen DA, Nielsen EM, Dasari T, Spellicy CJ. Pharmacogenetics of addiction therapy. Methods Mol Biol. 2014;1175:589–624.

Article   PubMed   CAS   Google Scholar  

Everitt BJ & Robbins TW. Drug addiction: updating actions to habits to compulsions ten years on. Annu Rev Psychol. 2015;67:23–50.

Article   PubMed   Google Scholar  

Belin D, Everitt BJ. Cocaine seeking habits depend upon dopamine-dependent serial connectivity linking the ventral with the dorsal striatum. Neuron. 2008;57:432–41.

Nestler EJ, Lüscher C. The molecular basis of drug addiction: linking epigenetic to synaptic and circuit mechanisms. Neuron. 2019;102:48–59.

Article   CAS   PubMed   PubMed Central   Google Scholar  

Nestler EJ. Cellular basis of memory for addiction. Dialogues Clin Neurosci. 2013;15:431–43.

Article   PubMed   PubMed Central   Google Scholar  

Nestler EJ. Molecular basis of long-term plasticity underlying addiction. Nat Rev Neurosci. 2001;2:119–28.

Anthony JC, Warner LA, Kessler RC. Comparative epidemiology of dependence on tobacco, alcohol, controlled substances, and inhalants: basic findings from the National Comorbidity Survey. Exp Clin Psychopharmacol. 1994;2:244–68.

Egervari G, Ciccocioppo R, Jentsch JD, Hurd YL. Shaping vulnerability to addiction – the contribution of behavior, neural circuits and molecular mechanisms. Neurosci Biobehav Rev. 2018;85:117–25.

Li MD, Burmeister M. New insights into the genetics of addiction. Nat. Rev. Genet. 2009;10:225–31.

Kreek MJ, Nielsen DA, Butelman ER, LaForge KS. Genetic influences on impulsivity, risk taking, stress responsivity and vulnerability to drug abuse and addiction. Nat Neurosci. 2005;8:1450–7.

Shohat S, Amelan A, Shifman S. Convergence and divergence in the genetics of psychiatric disorders from pathways to developmental stages. Biol Psychiatry. 2021;89:32–40.

Kandel, DB. Stages and Pathways of Drug Involvement: Examining the Gateway Hypothesis (Cambridge University Press, 2002).

Gerfen CR, Engber TM, Mahan LC, Susel Z, Chase TN, Monsma FJ Jr, et al. D1 and D2 dopamine receptor-regulated gene expression of striatonigral and striatopallidal neurons. Science. 1990;250:1429–32.

Schiffmann SN, Fisone G, Moresco R, Cunha RA, Ferré S. Adenosine A2A receptors and basal ganglia physiology. Prog. Neurobiol. 2007;83:277–92.

Gerfen CR, Surmeier DJ. Modulation of striatal projection systems by dopamine. Annu Rev Neurosci. 2011;34:441–66.

Di Chiara G, Imperato A. Drugs abused by humans preferentially increase synaptic dopamine concentrations in the mesolimbic system of freely moving rats. Proc Natl Acad Sci USA. 1988;85:5274–8.

Tanda G, Pontieri FE, Chiara GD. Cannabinoid and heroin activation of mesolimbic dopamine transmission by a common µ1 opioid receptor mechanism. Science. 1997;276:2048–50.

Pontieri FE, Tanda G, Orzi F, Chiara GD. Effects of nicotine on the nucleus accumbens and similarity to those of addictive drugs. Nature. 1996;382:255–7.

Nestler EJ. Is there a common molecular pathway for addiction? Nat Neurosci. 2005;8:1445–9.

Nestler EJ. The neurobiology of cocaine addiction. Sci Pract Perspect. 2005;3:4–10.

Kalivas PW, Volkow N, Seamans J. Unmanageable motivation in addiction: a pathology in prefrontal-accumbens glutamate transmission. Neuron. 2005;45:647–50.

Volkow ND, Fowler JS, Wang G-J, Swanson JM. Dopamine in drug abuse and addiction: results from imaging studies and treatment implications. Mol Psychiatry. 2004;9:557–69.

Chen SA, O'Dell LE, Hoefer ME, Greenwell TN, Zorrilla EP, Koob GF. Unlimited access to heroin self-administration: independent motivational markers of opiate dependence. Neuropsychopharmacology. 2006;31:2692–707.

Laviolette SR, Alexson TO, van der Kooy D. Lesions of the tegmental pedunculopontine nucleus block the rewarding effects and reveal the aversive effects of nicotine in the ventral tegmental area. J. Neurosci. 2002;22:8653–60.

Koob GF, Volkow ND. Neurocircuitry of addiction. Neuropsychopharmacology. 2010;35:217–38.

Pierce RC, Kumaresan V. The mesolimbic dopamine system: The final common pathway for the reinforcing effect of drugs of abuse? Neurosci Biobehav Rev. 2006;30:215–38.

Belin-Rauscent A, Fouyssac M, Bonci A, Belin D. How preclinical models evolved to resemble the diagnostic criteria of drug addiction. Biol. Psychiatry. 2016;79:39–46.

Piazza PV, Deroche-Gamonet V. A multistep general theory of transition to addiction. Psychopharmacology. 2013;229:387–413.

Pascoli V, Hiver A, Van Zessen R, Loureiro M, Achargui R, Harada M, et al. Stochastic synaptic plasticity underlying compulsion in a model of addiction. Nature. 2018;564:366–71.

Kauer JA, Malenka RC. Synaptic plasticity and addiction. Nat Rev Neurosci. 2007;8:844–58.

Mameli M, Balland B, Luján R, Lüscher C. Rapid synthesis and synaptic insertion of GluR2 for mGluR-LTD in the ventral tegmental area. Science. 2007;317:530–3.

Bellone C, Lüscher C. Cocaine triggered AMPA receptor redistribution is reversed in vivo by mGluR-dependent long-term depression. Nat Neurosci. 2006;9:636–41.

Pignatelli M, Bonci A. Role of dopamine neurons in reward and aversion: a synaptic plasticity perspective. Neuron. 2015;86:1145–57.

Stuber GD, Klanker M, de Ridder B, Bowers MS, Joosten RN, Feenstra MG, et al. Reward-predictive cues enhance excitatory synaptic strength onto midbrain dopamine neurons. Science. 2008;321:1690–2.

Bolam JP, Smith Y. The GABA and substance P input to dopaminergic neurones in the substantia nigra of the rat. Brain Res. 1990;529:57–78.

Henny P, Brown MT, Northrop A, Faunes M, Ungless MA, Magill PJ, et al. Structural correlates of heterogeneous in vivo activity of midbrain dopaminergic neurons. Nat Neurosci. 2012;15:613–9.

Bayer VE, Pickel VM. GABA-labeled terminals form proportionally more synapses with dopaminergic neurons containing low densities of tyrosine hydroxylase-immunoreactivity in rat ventral tegmental area. Brain Res. 1991;559:44–55.

Johnson SW, North RA. Two types of neurone in the rat ventral tegmental area and their synaptic inputs. J Physiol. 1992;450:455–68.

Edwards NJ, Tejeda HA, Pignatelli M, Zhang S, McDevitt RA, Wu J, et al. Circuit specificity in the inhibitory architecture of the VTA regulates cocaine-induced behavior. Nat Neurosci. 2017;20:438–48.

Menegas W, Bergan JF, Ogawa SK, Isogai Y, Umadevi Venkataraju K, Osten P, et al. Dopamine neurons projecting to the posterior striatum form an anatomically distinct subclass. eLife. 2015;4:e10032.

Ling W, Shoptaw S, Majewska D. Baclofen as a cocaine anti-craving medication: a preliminary clinical study. Neuropsychopharmacology. 1998;18:403–4.

CAS   PubMed   Google Scholar  

Young KA, Franklin TR, Roberts DC, Jagannathan K, Suh JJ, Wetherill RR, et al. Nipping cue reactivity in the bud: baclofen prevents limbic activation elicited by subliminal drug cues. J Neurosci. 2014;34:5038–43.

Article   PubMed   PubMed Central   CAS   Google Scholar  

Vocci FJ, Elkashef A. Pharmacotherapy and other treatments for cocaine abuse and dependence. Curr Opin Psychiatry. 2005;18:265–70.

Roberts DC, Andrews MM, Vickers GJ. Baclofen attenuates the reinforcing effects of cocaine in rats. Neuropsychopharmacology. 1996;15:417–23.

Slattery DA, Markou A, Froestl W, Cryan JF. The GABA B receptor-positive modulator GS39783 and the GABA B receptor agonist baclofen attenuate the reward-facilitating effects of cocaine: intracranial self-stimulation studies in the rat. Neuropsychopharmacology. 2005;30:2065–72.

Hotsenpiller G, Wolf ME. Baclofen attenuates conditioned locomotion to cues associated with cocaine administration and stabilizes extracellular glutamate levels in rat nucleus accumbens. Neuroscience. 2003;118:123–34.

Brebner K, Phelan R, Roberts DCS. Intra-VTA baclofen attenuates cocaine self-administration on a progressive ratio schedule of reinforcement. Pharmacol Biochem Behav. 2000;66:857–62.

Penn RD, Savoy SM, Corcos D, Latash M, Gottlieb G, Parke B, et al. Intrathecal baclofen for severe spinal spasticity. N Engl J Med. 1989;320:1517–21.

Sammaraiee Y, Yardley M, Keenan L, Buchanan K, Stevenson V, Farrell R. Intrathecal baclofen for multiple sclerosis related spasticity: a twenty year experience. Mult Scler Relat Disord. 2019;27:95–100.

Reynoard J, Schmitt C, Torrents R, Simon N. Toxicological considerations in the prescription of baclofen for the treatment of substance use disorders. Expert Opin Drug Metab Toxicol. 2020;16:309–17.

Garbutt JC, Kampov-Polevoy AB, Gallop R, Kalka-Juhl L, Flannery BA. Efficacy and safety of baclofen for alcohol dependence: a randomized, double-blind, placebo-controlled trial. Alcohol Clin Exp Res. 2010;34:1849–57.

Yamini D, Lee SH, Avanesyan A, Walter M, Runyon B. Utilization of baclofen in maintenance of alcohol abstinence in patients with alcohol dependence and alcoholic hepatitis with or without cirrhosis. Alcohol Alcohol. 2014;49:453–6.

Addolorato G, Leggio L, Hopf FW, Diana M, Bonci A. Novel therapeutic strategies for alcohol and drug addiction: focus on GABA, ion channels and transcranial magnetic stimulation. Neuropsychopharmacology. 2012;37:163–77.

Kahn R, Biswas K, Childress AR, Shoptaw S, Fudala PJ, Gorgon L, et al. Multi-center trial of baclofen for abstinence initiation in severe cocaine-dependent individuals. Drug Alcohol Depend. 2009;103:59–64.

Dore GM, Lo K, Juckes L, Bezyan S, Latt N. Clinical experience with baclofen in the management of alcohol-dependent patients with psychiatric comorbidity: a selected case series. Alcohol Alcohol. 2011;46:714–20.

Nugent FS, Penick EC, Kauer JA. Opioids block long-term potentiation of inhibitory synapses. Nature. 2007;446:1086–90.

Liu Q, Pu L, Poo M. Repeated cocaine exposure in vivo facilitates LTP induction in midbrain dopamine neurons. Nature. 2005;437:1027–31.

Barrett AC, Negus SS, Mello NK, Caine SB. Effect of GABA agonists and GABA-A receptor modulators on cocaine- and food-maintained responding and cocaine discrimination in rats. J Pharmacol Exp Ther. 2005;315:858–71.

Brodie JD, Figueroa E, Dewey SL. Treating cocaine addiction: from preclinical to clinical trial experience with γ-vinyl GABA. Synapse. 2003;50:261–5.

Barker JS, Hines RM. Regulation of GABAA receptor subunit expression in substance use disorders. Int J Mol Sci. 2020;21:4445.

Article   CAS   PubMed Central   Google Scholar  

Tan KR, Rudolph U, Lüscher C. Hooked on benzodiazepines: GABAA receptor subtypes and addiction. Trends Neurosci. 2011;34:188–97.

Brands B, Blake J, Marsh DC, Sproule B, Jeyapalan R, Li S. The impact of benzodiazepine use on methadone maintenance treatment outcomes. J Addict Dis. 2008;27:37–48.

Dacher M, Nugent FS. Morphine-induced modulation of LTD at GABAergic synapses in the ventral tegmental area. Neuropharmacology. 2011;61:1166–71.

Authement ME, Langlois LD, Kassis H, Gouty S, Dacher M, Shepard RD, et al. Morphine-induced synaptic plasticity in the VTA is reversed by HDAC inhibition. J Neurophysiol. 2016;116:1093–103.

Abraham WC, Bear MF. Metaplasticity: the plasticity of synaptic plasticity. Trends Neurosci. 1996;19:126–30.

Martin M, Chen BT, Hopf FW, Bowers MS, Bonci A. Cocaine self-administration selectively abolishes LTD in the core of the nucleus accumbens. Nat. Neurosci. 2006;9:868–9.

Thomas MJ, Beurrier C, Bonci A, Malenka RC. Long-term depression in the nucleus accumbens: a neural correlate of behavioral sensitization to cocaine. Nat. Neurosci. 2001;4:1217–23.

Boudreau AC, Wolf ME. Behavioral sensitization to cocaine is associated with increased AMPA receptor surface expression in the nucleus accumbens. J. Neurosci. 2005;25:9144–51.

Ferrario CR, Li X, Wang X, Reimers JM, Uejima JL, Wolf ME. The role of glutamate receptor redistribution in locomotor sensitization to cocaine. Neuropsychopharmacology. 2010;35:818–33.

Conrad KL, Tseng KY, Uejima JL, Reimers JM, Heng LJ, Shaham Y, et al. Formation of accumbens GluR2-lacking AMPA receptors mediates incubation of cocaine craving. Nature. 2008;454:118–21. https://doi.org/10.1038/nature06995 .

Wolf ME, Tseng KY. Calcium-permeable AMPA receptors in the VTA and nucleus accumbens after cocaine exposure:when, how, and why? Front Mol Neurosci. 2012;5:72.

Grimm JW, Hope BT, Wise RA, Shaham Y. Neuroadaptation. Incubation of cocaine craving after withdrawal. Nature. 2001;412:141–2.

Sadler-Smith E. Wallas’ four-stage model of the creative process: more than meets the eye? Creat Res J. 2015;27:342–52.

Gawin FH, Kleber HD. Abstinence symptomatology and psychiatric diagnosis in cocaine abusers. Clinical observations. Arch Gen Psychiatry. 1986;43:107–13.

Childress AR, Mozley PD, McElgin W, Fitzgerald J, Reivich M, O'Brien CP. Limbic activation during cue-induced cocaine craving. Am J Psychiatry. 1999;156:11–18.

Parvaz MA, Moeller SJ, Goldstein RZ. Incubation of cue-induced craving in adults addicted to cocaine measured by electroencephalography. JAMA Psychiatry. 2016;73:1127–34.

Lippman-Bell JJ, Zhou C, Sun H, Feske JS, Jensen FE. Early-life seizures alter synaptic calcium-permeable AMPA receptor function and plasticity. Mol Cell Neurosci. 2016;76:11–20.

Achuta VS, Möykkynen T, Peteri UK, Turconi G, Rivera C, Keinänen K, et al. Functional changes of AMPA responses in human induced pluripotent stem cell-derived neural progenitors in fragile X syndrome. Sci Sÿignal. 2018;11:eaan8784.

Article   CAS   Google Scholar  

Selvaraj BT, Livesey MR, Zhao C, Gregory JM, James OT, Cleary EM, et al. C9ORF72 repeat expansion causes vulnerability of motor neurons to Ca 2+ -permeable AMPA receptor-mediated excitotoxicity. Nat Commun. 2018;9:347.

Kobylecki C, Cenci MA, Crossman AR, Ravenscroft P. Calcium-permeable AMPA receptors are involved in the induction and expression of l-DOPA-induced dyskinesia in Parkinson’s disease. J Neurochem. 2010;114:499–511.

Whitehead G, Regan P, Whitcomb DJ, Cho K. Ca2+-permeable AMPA receptor: a new perspective on amyloid-beta mediated pathophysiology of Alzheimer’s disease. Neuropharmacology. 2017;112:221–7.

Twomey EC, Yelshanskaya MV, Vassilevski AA, Sobolevsky AI. Mechanisms of channel block in calcium-permeable AMPA receptors. Neuron. 2018;99:956–.e4.

Loweth JA, Scheyer AF, Milovanovic M, LaCrosse AL, Flores-Barrera E, Werner CT, et al. Synaptic depression via mGluR1 positive allosteric modulation suppresses cue-induced cocaine craving. Nat. Neurosci. 2014;17:73–80.

Scheyer AF, Loweth JA, Christian DT, Uejima J, Rabei R, Le T, et al. AMPA receptor plasticity in accumbens core contributes to incubation of methamphetamine craving. Biol. Psychiatry. 2016;80:661–70.

Pascoli V, Terrier J, Espallergues J, Valjent E, O'Connor EC, Lüscher C. Contrasting forms of cocaine-evoked plasticity control components of relapse. Nature. 2014;509:459–64.

Ma T, Cheng Y, Roltsch Hellard E, Wang X, Lu J, Gao X, et al. Bidirectional and long-lasting control of alcohol-seeking behavior by corticostriatal LTP and LTD. Nat. Neurosci. 2018;21:373–83.

Pascoli V, Turiault M, Lüscher C. Reversal of cocaine-evoked synaptic potentiation resets drug-induced adaptive behaviour. Nature. 2012;481:71–75.

Valjent E, Bertran-Gonzalez J, Aubier B, Greengard P, Hervé D, Girault JA. Mechanisms of locomotor sensitization to drugs of abuse in a two-injection protocol. Neuropsychopharmacology 2010;35:401–15.

Robinson TE, Berridge KC. The neural basis of drug craving: an incentive-sensitization theory of addiction. Brain Res Brain Res Rev. 1993;18:247–91.

Schenk S, Partridge B. Sensitization and tolerance in psychostimulant self-administration. Pharmacol Biochem Behav. 1997;57:543–50.

Vezina P, Lorrain DS, Arnold GM, Austin JD, Suto N. Sensitization of midbrain dopamine neuron reactivity promotes the pursuit of amphetamine. J Neurosci. 2002;22:4654–62.

De Vries TJ, Schoffelmeer AN, Binnekade R, Mulder AH, Vanderschuren LJ. Drug-induced reinstatement of heroin- and cocaine-seeking behaviour following long-term extinction is associated with expression of behavioural sensitization. Eur J Neurosci. 1998;10:3565–71.

Suto N, Tanabe LM, Austin JD, Creekmore E, Pham CT, Vezina P. Previous exposure to psychostimulants enhances the reinstatement of cocaine seeking by nucleus accumbens AMPA. Neuropsychopharmacology. 2004;29:2149–59.

Vorspan F, de Witt P, Zerdazi EH, Karsinti E, Ksouda K, Icick R, et al. Chronic exposure to cocaine is associated with persistent behavioral disturbances. A cross-sectional dimensional study in outpatients with multiple substance use disorders. Psychopharmacology. 2020;237:3399–407.

Vorspan F, Icick R, Mekdad N, Courtin C, Bloch V, Bellivier F, et al. Translational study of the whole transcriptome in rats and genetic polymorphisms in humans identifies LRP1B and VPS13A as key genes involved in tolerance to cocaine-induced motor disturbances. Transl Psychiatry. 2020;10:381.

Vanderschuren LJ, Kalivas PW. Alterations in dopaminergic and glutamatergic transmission in the induction and expression of behavioral sensitization: a critical review of preclinical studies. Psychopharmacology. 2000;151:99–120.

Vezina P, Leyton M. Conditioned cues and the expression of stimulant sensitization in animals and humans. Neuropharmacology. 2009;56:160–8.

Tzschentke TM. REVIEW ON CPP: measuring reward with the conditioned place preference (CPP) paradigm: update of the last decade. Addict Biol. 2007;12:227–462.

Bardo MT, Bevins RA. Conditioned place preference: what does it add to our preclinical understanding of drug reward? Psychopharmacology. 2000;153:31–43.

Itzhak Y, Martin JL. Cocaine-induced conditioned place preference in mice: induction, extinction and reinstatement by related psychostimulants. Neuropsychopharmacology. 2002;26:130–4.

Creed M, Pascoli VJ, Lüscher C. Refining deep brain stimulation to emulate optogenetic treatment of synaptic pathology. Science. 2015;347:659–64.

Shen Y, Campbell RE, Côté DC, Paquet M-E. Challenges for therapeutic applications of opsin-based optogenetic tools in humans. Front Neural Circuits. 2020;14:41.

Vorspan F, Mallet L, Corvol J-C, Pelissolo A, Lépine J-P. Treating addictions with deep brain stimulation is premature but well-controlled clinical trials should be performed. Addict Abingdon Engl. 2011;106:1535–6.

Mahoney JJ, Hanlon CA, Marshalek PJ, Rezai AR, Krinke L. Transcranial magnetic stimulation, deep brain stimulation, and other forms of neuromodulation for substance use disorders: review of modalities and implications for treatment. J Neurol Sci. 2020;418:117149.

Shen W, Flajolet M, Greengard P, Surmeier DJ. Dichotomous dopaminergic control of striatal synaptic plasticity. Science. 2008;321:848–51.

Moussawi K, Pacchioni A, Moran M, Olive MF, Gass JT, Lavin A, et al. N -Acetylcysteine reverses cocaine-induced metaplasticity. Nat Neurosci. 2009;12:182–9.

Gerfen CR. The neostriatal mosaic: multiple levels of compartmental organization. Trends Neurosci. 1992;15:133–9.

Vicente AM, Galvão-Ferreira P, Tecuapetla F, Costa RM. Direct and indirect dorsolateral striatum pathways reinforce different action strategies. Curr Biol. 2016;26:R267–269.

Freeze BS, Kravitz AV, Hammack N, Berke JD, Kreitzer AC. Control of basal ganglia output by direct and indirect pathway projection neurons. J Neurosci. 2013;33:18531–9.

Durieux PF, Schiffmann SN, de Kerchove d’Exaerde A. Differential regulation of motor control and response to dopaminergic drugs by D1R and D2R neurons in distinct dorsal striatum subregions. EMBO J. 2012;31:640–53.

Cox J, Witten IB. Striatal circuits for reward learning and decision-making. Nat Rev Neurosci. 2019;20:482–94.

Lalchandani RR, van der Goes M-S, Partridge JG, Vicini S. Dopamine D2 receptors regulate collateral inhibition between striatal medium spiny neurons. J Neurosci. 2013;33:14075–86.

Dobbs LK, Kaplan AR, Lemos JC, Matsui A, Rubinstein M, Alvarez VA. Dopamine regulation of lateral inhibition between striatal neurons gates the stimulant actions of cocaine. Neuron. 2016;90:1100–13.

Dobbs LK, Kaplan AR, Bock R, Phamluong K, Shin JH, Bocarsly ME, et al. D1 receptor hypersensitivity in mice with low striatal D2 receptors facilitates select cocaine behaviors. Neuropsychopharmacology. 2019;44:805–16.

Tecuapetla F, Koós T, Tepper JM, Kabbani N, Yeckel MF. Differential dopaminergic modulation of neostriatal synaptic connections of striatopallidal axon collaterals. J Neurosci. 2009;29:8977–90.

Mulholland PJ, Chandler LJ, Kalivas PW. Signals from the fourth dimension regulate drug relapse. Trends Neurosci. 2016;39:472–85.

Smith AC, Kupchik YM, Scofield MD, Gipson CD, Wiggins A, Thomas CA, et al. Synaptic plasticity mediating cocaine relapse requires matrix metalloproteinases. Nat Neurosci. 2014;17:1655–7.

Moussawi K, Zhou W, Shen H, Reichel CM, See RE, Carr DB, et al. Reversing cocaine-induced synaptic potentiation provides enduring protection from relapse. Proc Natl Acad Sci USA. 2011;108:385–90.

Abulseoud OA, Camsari UM, Ruby CL, Kasasbeh A, Choi S, Choi DS. Attenuation of ethanol withdrawal by ceftriaxone-induced upregulation of glutamate transporter EAAT2. Neuropsychopharmacology. 2014;39:1674–84.

Knackstedt LA, Melendez RI, Kalivas PW. Ceftriaxone restores glutamate homeostasis and prevents relapse to cocaine seeking. Biol Psychiatry. 2010;67:81–84.

Huntley GW. Synaptic circuit remodelling by matrix metalloproteinases in health and disease. Nat. Rev. Neurosci. 2012;13:743–57.

Brown TE, Forquer MR, Harding JW, Wright JW, Sorg BA. Increase in matrix metalloproteinase-9 levels in the rat medial prefrontal cortex after cocaine reinstatement of conditioned place preference. Synapse. 2008;62:886–9.

Mizoguchi H, Yamada K, Mouri A, Niwa M, Mizuno T, Noda Y, et al. Role of matrix metalloproteinase and tissue inhibitor of MMP in methamphetamine-induced behavioral sensitization and reward: implications for dopamine receptor down-regulation and dopamine release. J Neurochem. 2007;102:1548–60.

Kroener S, Mulholland PJ, New NN, Gass JT, Becker HC, Chandler LJ. Chronic alcohol exposure alters behavioral and synaptic plasticity of the rodent prefrontal cortex. PLoS ONE. 2012;7:e37541.

Nimitvilai S, Lopez MF, Mulholland PJ, Woodward JJ. Chronic intermittent ethanol exposure enhances the excitability and synaptic plasticity of lateral orbitofrontal cortex neurons and induces a tolerance to the acute inhibitory actions of ethanol. Neuropsychopharmacology. 2016;41:1112–27.

Kasanetz F, Lafourcade M, Deroche-Gamonet V, Revest JM, Berson N, Balado E, et al. Prefrontal synaptic markers of cocaine addiction-like behavior in rats. Mol Psychiatry. 2013;18:729–37.

Meinhardt MW, Hansson AC, Perreau-Lenz S, Bauder-Wenz C, Stählin O, Heilig M, et al. Rescue of infralimbic mGluR2 deficit restores control over drug-seeking behavior in alcohol dependence. J Neurosci. 2013;33:2794–806.

Lu L, Uejima JL, Gray SM, Bossert JM, Shaham Y. Systemic and central amygdala injections of the mGluR2/3 agonist LY379268 attenuate the expression of incubation of cocaine craving. Biol Psychiatry. 2007;61:591–8.

Baptista MAS, Martin-Fardon R, Weiss F. Preferential effects of the metabotropic glutamate 2/3 receptor agonist LY379268 on conditioned reinstatement versus primary reinforcement: comparison between cocaine and a potent conventional reinforcer. J Neurosci. 2004;24:4723–7.

Peters J, Kalivas PW. The group II metabotropic glutamate receptor agonist, LY379268, inhibits both cocaine- and food-seeking behavior in rats. Psychopharmacology. 2006;186:143–9.

Caprioli D, Justinova Z, Venniro M, Shaham Y. Effect of novel allosteric modulators of metabotropic glutamate receptors on drug self-administration and relapse: a review of preclinical studies and their clinical implications. Biol Psychiatry. 2018;84:180–92.

Clark AM et al. Dopamine D2 receptors in the paraventricular thalamus attenuate cocaine locomotor sensitization. eNeuro. 2017;4:ENEURO.0227-17.2017.

Dong X, Li S, Kirouac GJ. Collateralization of projections from the paraventricular nucleus of the thalamus to the nucleus accumbens, bed nucleus of the stria terminalis, and central nucleus of the amygdala. Brain Struct. Funct. 2017;222:3927–43.

Huang AS, Mitchell JA, Haber SN, Alia-Klein N & Goldstein RZ. The thalamus in drug addiction: from rodents to humans. Philos Trans R Soc Lond B Biol Sci. 2018;373:20170028.

Perez SM, Lodge DJ. Convergent inputs from the hippocampus and thalamus to the nucleus accumbens regulate dopamine neuron activity. J Neurosci. 2018;38:10607–18.

Kelley AE, Baldo BA, Pratt WE. A proposed hypothalamic–thalamic–striatal axis for the integration of energy balance, arousal, and food reward. J Comp Neurol. 2005;493:72–85.

Penzo MA & Gao C. The paraventricular nucleus of the thalamus: an integrative node underlying homeostatic behavior. Trends Neurosci. 2021. https://doi.org/10.1016/j.tins.2021.03.001 .

De Groote A & de Kerchove d'Exaerde A. Thalamo-Nucleus Accumbens projections in motivated behaviors and addiction. Front Syst Neurosci. 2021;15:711350.

Zhu Y, Wienecke CFR, Nachtrab G, Chen X. A thalamic input to the nucleus accumbens mediates opiate dependence. Nature. 2016;530:219–22.

Chisholm A, Rizzo D, Fortin É, Moman V, Quteishat N, Romano A, et al. Assessing the role of corticothalamic and thalamo-accumbens projections in the augmentation of heroin seeking in chronically food-restricted rats. J Neurosci. 2021;41:354–65.

Otis JM, Zhu M, Namboodiri V, Cook CA, Kosyk O, Matan AM, et al. Paraventricular thalamus projection neurons integrate cortical and hypothalamic signals for cue-reward processing. Neuron. 2019;103:423–.e4.

Giros B, Jaber M, Jones SR, Wightman RM, Caron MG. Hyperlocomotion and indifference to cocaine and amphetamine in mice lacking the dopamine transporter. Nature. 1996;379:606–12.

Sora I, Hall FS, Andrews AM, Itokawa M, Li XF, Wei HB, et al. Molecular mechanisms of cocaine reward: combined dopamine and serotonin transporter knockouts eliminate cocaine place preference. Proc Natl Acad Sci USA. 2001;98:5300–5.

Yang H-J, Zhang HY, Bi GH, He Y, Gao JT, Xi ZX. Deletion of type 2 metabotropic glutamate receptor decreases sensitivity to cocaine reward in rats. Cell Rep. 2017;20:319–32.

Chiamulera C, Epping-Jordan MP, Zocchi A, Marcon C, Cottiny C, Tacconi S, et al. Reinforcing and locomotor stimulant effects of cocaine are absent in mGluR5 null mutant mice. Nat Neurosci. 2001;4:873–4.

De Backer J-F, et al. Deletion of Maged1 in mice abolishes locomotor and reinforcing effects of cocaine. EMBO Rep. 2018;19:e45089.

Karadurmus D, Rial D, De Backer JF, Communi D, de Kerchove d'Exaerde A, Schiffmann SN. GPRIN3 controls neuronal excitability, morphology, and striatal-dependent behaviors in the indirect pathway of the striatum. J. Neurosci. 2019;39:7513–28.

Renthal W, Kumar A, Xiao G, Wilkinson M, Covington HE, Maze I, et al. Genome-wide analysis of chromatin regulation by cocaine reveals a role for sirtuins. Neuron. 2009;62:335–48.

Ena SL, De Backer J-F, Schiffmann SN, de Kerchove d’Exaerde A. FACS array profiling identifies Ecto-5’ nucleotidase as a striatopallidal neuron-specific gene involved in striatal-dependent learning. J Neurosci. 2013;33:8794–809.

Iida, N & Kozasa, T. 29 - Identification and biochemical analysis of GRIN1 and GRIN2. In: Siderovski DP, editor. Methods in Enzymology. Academic Press; 2004;390:475–83.

Mototani Y, Okamura T, Goto M, Shimizu Y, Yanobu-Takanashi R, Ito A, et al. Role of G protein-regulated inducer of neurite outgrowth 3 (GRIN3) in β-arrestin 2-Akt signaling and dopaminergic behaviors. Pflügers Arch. 2018;470:937–47.

Ventura A, Meissner A, Dillon CP, McManus M, Sharp PA, Van Parijs L, et al. Cre-lox-regulated conditional RNA interference from transgenes. Proc Natl Acad Sci USA. 2004;101:10380–5.

David Smith A, Paul Bolam J. The neural network of the basal ganglia as revealed by the study of synaptic connections of identified neurones. Trends Neurosci. 1990;13:259–65.

Robinson TE, Kolb B. Alterations in the morphology of dendrites and dendritic spines in the nucleus accumbens and prefrontal cortex following repeated treatment with amphetamine or cocaine. Eur J Neurosci. 1999;11:1598–604.

Spencer S, Garcia-Keller C, Roberts-Wolfe D, Heinsbroek JA, Mulvaney M, Sorrell A, et al. Cocaine use reverses striatal plasticity produced during cocaine seeking. Biol Psychiatry. 2017;81:616–24.

MacAskill AF, Cassel JM, Carter AG. Cocaine exposure reorganizes cell type– and input-specific connectivity in the nucleus accumbens. Nat Neurosci. 2014;17:1198–207.

Dos Santos M, Cahill EN, Bo GD, Vanhoutte P, Caboche J, Giros B, et al. Cocaine increases dopaminergic connectivity in the nucleus accumbens. Brain Struct Funct. 2018;223:913–23.

Jaenisch R, Bird A. Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals. Nat Genet. 2003;33:245–54.

Nestler EJ. Epigenetic mechanisms of drug addiction. Neuropharmacology. 2014;76:259–68.

Hamilton PJ, Nestler EJ. Epigenetics and addiction. Curr Opin Neurobiol. 2019;59:128–36.

Handel AE, Ramagopalan SV. Is Lamarckian evolution relevant to medicine? BMC Med Genet. 2010;11:73.

Jablonka E & Lamb MJ. Epigenetic Inheritance And Evolution: The Lamarckian Dimension. Oxford University Press; 1999.

Springer JT & Holley D. An Introduction To Zoology: Investigating The Animal World. Jones & Bartlett Learning; 2013.

Vassoler FM, White SL, Schmidt HD, Sadri-Vakili G, Pierce RC. Epigenetic inheritance of a cocaine-resistance phenotype. Nat. Neurosci. 2013;16:42–47.

Goldberg LR, Gould TJ. Multigenerational and transgenerational effects of paternal exposure to drugs of abuse on behavioral and neural function. Eur J Neurosci. 2019;50:2453–66.

Sadri-Vakili G, Kumaresan V, Schmidt HD, Famous KR, Chawla P, Vassoler FM, et al. Cocaine-induced chromatin remodeling increases brain-derived neurotrophic factor transcription in the rat medial prefrontal cortex, which alters the reinforcing efficacy of cocaine. J Neurosci. 2010;30:11735–44.

Schmidt HD, Sangrey GR, Darnell SB, Schassburger RL, Cha JH, Pierce RC, et al. Increased brain-derived neurotrophic factor (BDNF) expression in the ventral tegmental area during cocaine abstinence is associated with increased histone acetylation at BDNF exon I-containing promoters. J Neurochem. 2012;120:202–9.

Gavin DP, et al. Stable histone methylation changes at proteoglycan network genes following ethanol exposure. Front Genet. 2018;9:346.

Sadakierska-Chudy A, Frankowska M, Jastrzębska J, Wydra K, Miszkiel J, Sanak M, et al. Cocaine administration and its withdrawal enhance the expression of genes encoding histone-modifying enzymes and histone acetylation in the rat prefrontal cortex. Neurotox Res. 2017;32:1–10.

Allis CD, Jenuwein T. The molecular hallmarks of epigenetic control. Nat Rev Genet. 2016;17:487–500.

Wang L, Lv Z, Hu Z, Sheng J, Hui B, Sun J, et al. Chronic cocaine-induced H3 acetylation and transcriptional activation of CaMKIIα in the nucleus accumbens is critical for motivation for drug reinforcement. Neuropsychopharmacology. 2010;35:913–28.

Brami-Cherrier K. Parsing molecular and behavioral effects of cocaine in mitogen- and stress-activated protein kinase-1-deficient mice. J Neurosci. 2005;25:11444–54.

Barrett RM, Wood MA. Beyond transcription factors: the role of chromatin modifying enzymes in regulating transcription required for memory. Learn Mem. 2008;15:460–7.

Malvaez M, Mhillaj E, Matheos DP, Palmery M, Wood MA. CBP in the nucleus accumbens regulates cocaine-induced histone acetylation and is critical for cocaine-associated behaviors. J Neurosci. 2011;31:16941–8.

Tsankova NM, Berton O, Renthal W, Kumar A, Neve RL, Nestler EJ. Sustained hippocampal chromatin regulation in a mouse model of depression and antidepressant action. Nat Neurosci. 2006;9:519–25.

Kumar A, Choi KH, Renthal W, Tsankova NM, Theobald DE, Truong HT, et al. Chromatin remodeling is a key mechanism underlying cocaine-induced plasticity in striatum. Neuron. 2005;48:303–14.

Sun J, Wang L, Jiang B, Hui B, Lv Z, Ma L. The effects of sodium butyrate, an inhibitor of histone deacetylase, on the cocaine- and sucrose-maintained self-administration in rats. Neurosci Lett. 2008;441:72–76.

Romieu P, Host L, Gobaille S, Sandner G, Aunis D, Zwiller J. Histone deacetylase inhibitors decrease cocaine but not sucrose self-administration in rats. J. Neurosci. 2008;28:9342–8.

Levine AA, Guan Z, Barco A, Xu S, Kandel ER, Schwartz JH. CREB-binding protein controls response to cocaine by acetylating histones at the fosB promoter in the mouse striatum. Proc Natl Acad Sci USA. 2005;102:19186–91.

Kalda A, Heidmets L-T, Shen H-Y, Zharkovsky A, Chen J-F. Histone deacetylase inhibitors modulates the induction and expression of amphetamine-induced behavioral sensitization partially through an associated learning of the environment in mice. Behav. Brain Res. 2007;181:76–84.

Kennedy PJ, Feng J, Robison AJ, Maze I, Badimon A, Mouzon E, et al. Class I HDAC inhibition blocks cocaine-induced plasticity by targeted changes in histone methylation. Nat Neurosci. 2013;16:434–40.

LaPlant Q, Vialou V, Covington HE, Dumitriu D, Feng J, Warren BL, et al. Dnmt3a regulates emotional behavior and spine plasticity in the nucleus accumbens. Nat Neurosci. 2010;13:1137–43.

Massart R, Barnea R, Dikshtein Y, Suderman M, Meir O, Hallett M, et al. Role of DNA methylation in the nucleus accumbens in incubation of cocaine craving. J Neurosci. 2015;35:8042–58.

Bertran-Gonzalez J, Bosch C, Maroteaux M, Matamales M, Hervé D, Valjent E, et al. Opposing patterns of signaling activation in dopamine D1 and D2 receptor-expressing striatal neurons in response to cocaine and haloperidol. J Neurosci. 2008;28:5671–85.

Brami‐Cherrier K, Roze E, Girault J-A, Betuing S, Caboche J. Role of the ERK/MSK1 signalling pathway in chromatin remodelling and brain responses to drugs of abuse. J Neurochem. 2009;108:1323–35.

Scobie KN, Damez-Werno D, Sun H, Shao N, Gancarz A, Panganiban CH, et al. Essential role of poly(ADP-ribosyl)ation in cocaine action. Proc Natl Acad Sci USA. 2014;111:2005–10.

Lepack AE, Werner CT, Stewart AF, Fulton SL, Zhong P, Farrelly LA, et al. Dopaminylation of histone H3 in ventral tegmental area regulates cocaine seeking. Science. 2020;368:197–201.

Sun H, Maze I, Dietz DM, Scobie KN, Kennedy PJ, Damez-Werno D, et al. Morphine epigenomically regulates behavior through alterations in histone H3 lysine 9 dimethylation in the nucleus accumbens. J Neurosci. 2012;32:17454–64.

Maze I, Feng J, Wilkinson MB, Sun H, Shen L, Nestler EJ. Cocaine dynamically regulates heterochromatin and repetitive element unsilencing in nucleus accumbens. Proc Natl Acad Sci USA. 2011;108:3035–40.

Liu XS, Wu H, Ji X, Stelzer Y, Wu X, Czauderna S, et al. Editing DNA methylation in the mammalian genome. Cell. 2016;167:233–247.e17.

Liao H-K, Hatanaka F, Araoka T, Reddy P, Wu MZ, Sui Y, et al. In vivo target gene activation via CRISPR/Cas9-mediated trans-epigenetic modulation. Cell. 2017;171:1495–1507.e15.

Heller EA, Cates HM, Peña CJ, Sun H, Shao N, Feng J, et al. Locus-specific epigenetic remodeling controls addiction- and depression-related behaviors. Nat. Neurosci. 2014;17:1720–7.

Heller EA, Hamilton PJ, Burek DD, Lombroso SI, Peña CJ, Neve RL, et al. Targeted epigenetic remodeling of the Cdk5 gene in nucleus accumbens regulates cocaine- and stress-evoked behavior. J Neurosci. 2016;36:4690–7.

Monsey MS, Ota KT, Akingbade IF, Hong ES, Schafe GE. Epigenetic alterations are critical for fear memory consolidation and synaptic plasticity in the lateral amygdala. PLoS ONE. 2011;6:e19958.

Massart R, Freyburger M, Suderman M, Paquet J, El Helou J, Belanger-Nelson E, et al. The genome-wide landscape of DNA methylation and hydroxymethylation in response to sleep deprivation impacts on synaptic plasticity genes. Transl Psychiatry. 2014;4:e347–e347.

Kyzar EJ, Pandey SC. Molecular mechanisms of synaptic remodeling in alcoholism. Neurosci Lett. 2015;601:11–19.

Rogge GA, Wood MA. The role of histone acetylation in cocaine-induced neural plasticity and behavior. Neuropsychopharmacology. 2013;38:94–110.

Kyzar EJ, Floreani C, Teppen TL & Pandey SC. Adolescent alcohol exposure: burden of epigenetic reprogramming, synaptic remodeling, and adult psychopathology. Front. Neurosci. 2016;10:222.

Maze I, Covington HE, Dietz DM, LaPlant Q, Renthal W, Russo SJ, et al. Essential role of the histone methyltransferase G9a in cocaine-induced plasticity. Science. 2010;327:213–6.

Robison AJ, Nestler EJ. Transcriptional and epigenetic mechanisms of addiction. Nat Rev Neurosci. 2011;12:623–37.

Lobo MK, Zaman S, Damez-Werno DM, Koo JW, Bagot RC, DiNieri JA, et al. ΔFosB induction in striatal medium spiny neuron subtypes in response to chronic pharmacological, emotional, and optogenetic stimuli. J Neurosci. 2013;33:18381–95.

Huang YH, Lin Y, Brown TE, Han MH, Saal DB, Neve RL, et al. CREB modulates excitability of nucleus accumbens neurons. Nat Neurosci. 2006;9:475–7.

Murphy DD, Segal M. Morphological plasticity of dendritic spines in central neurons is mediated by activation of cAMP response element binding protein. Proc Natl Acad Sci USA. 1997;94:1482–7.

Grueter BA, Robison AJ, Neve RL, Nestler EJ, Malenka RC. ∆FosB differentially modulates nucleus accumbens direct and indirect pathway function. Proc Natl Acad Sci USA. 2013;110:1923–8.

Smith ACW, Kenny PJ. MicroRNAs regulate synaptic plasticity underlying drug addiction. Genes Brain Behav. 2018;17:e12424.

Nudelman AS, DiRocco DP, Lambert TJ, Garelick MG, Le J, Nathanson NM, et al. Neuronal activity rapidly induces transcription of the CREB-regulated microRNA-132, in vivo. Hippocampus. 2010;20:492–8.

CAS   PubMed   PubMed Central   Google Scholar  

Yasui DH, Peddada S, Bieda MC, Vallero RO, Hogart A, Nagarajan RP, et al. Integrated epigenomic analyses of neuronal MeCP2 reveal a role for long-range interaction with active genes. Proc Natl Acad Sci USA. 2007;104:19416–21.

Chahrour M, Jung SY, Shaw C, Zhou X, Wong ST, Qin J, et al. MeCP2, a key contributor to neurological disease, activates and represses transcription. Science. 2008;320:1224–9.

De Crescenzo F, Ciabattini M, D'Alò GL, De Giorgi R, Del Giovane C, Cassar C, et al. Comparative efficacy and acceptability of psychosocial interventions for individuals with cocaine and amphetamine addiction: a systematic review and network meta-analysis. PLOS Med. 2018;15:e1002715.

Phillips KA, Epstein DH, Preston KL. Psychostimulant addiction treatment. Neuropharmacology. 2014;87:150–60.

Mattick RP, Breen C, Kimber J & Davoli M. Methadone maintenance therapy versus no opioid replacement therapy for opioid dependence. Cochrane Database Syst. Rev. 2009;CD002209. https://doi.org/10.1002/14651858.CD002209.pub2 .

Hartmann‐Boyce J, Chepkin SC, Ye W, Bullen C & Lancaster T. Nicotine replacement therapy versus control for smoking cessation. Cochrane Database Syst Rev. 2018. https://doi.org/10.1002/14651858.CD000146.pub5 .

Tardelli VS, Bisaga A, Arcadepani FB, Gerra G, Levin FR, Fidalgo TM. Prescription psychostimulants for the treatment of stimulant use disorder: a systematic review and meta-analysis. Psychopharmacology. 2020;237:2233–55.

Darke S & Farrell M. Which medications are suitable for agonist drug maintenance? Addiction. 2016;111:767–74.

Kosten TR, Domingo CB, Shorter D, Orson F, Green C, Somoza E, et al. Vaccine for cocaine dependence: a randomized double-blind placebo-controlled efficacy trial. Drug Alcohol Depend. 2014;140:42–47.

Hossain MK, Hassanzadeganroudsari M, Nurgali K, Apostolopoulos V. Vaccine development against methamphetamine drug addiction. Expert Rev Vaccines. 2020;19:1105–14.

Dackis C, O’Brien C. Neurobiology of addiction: treatment and public policy ramifications. Nat Neurosci. 2005;8:1431–6.

Benabid AL, Pollak P, Gervason C, Hoffmann D, Gao DM, Hommel M, et al. Long-term suppression of tremor by chronic stimulation of the ventral intermediate thalamic nucleus. Lancet. 1991;337:403–6.

Bergman H, Wichmann T, DeLong MR. Reversal of experimental parkinsonism by lesions of the subthalamic nucleus. Science. 1990;249:1436–8.

Limousin P, Pollak P, Benazzouz A, Hoffmann D, Le Bas JF, Broussolle E, et al. Effect on parkinsonian signs and symptoms of bilateral subthalamic nucleus stimulation. Lancet. 1995;345:91–95.

Johansen-Berg H, Gutman DA, Behrens TE, Matthews PM, Rushworth MF, Katz E, et al. Anatomical connectivity of the subgenual cingulate region targeted with deep brain stimulation for treatment-resistant depression. Cereb Cortex. 2008;1991:1374–83.

Greenberg BD, Malone DA, Friehs GM, Rezai AR, Kubu CS, Malloy PF, et al. Three-year outcomes in deep brain stimulation for highly resistant obsessive-compulsive disorder. Neuropsychopharmacology. 2006;31:2384–93.

Porta M, Brambilla A, Cavanna AE, Servello D, Sassi M, Rickards H, et al. Thalamic deep brain stimulation for treatment-refractory Tourette syndrome: two-year outcome. Neurology. 2009;73:1375–80.

Kuhn J, Lenartz D, Huff W, Lee S, Koulousakis A, Klosterkoetter J, et al. Remission of alcohol dependency following deep brain stimulation of the nucleus accumbens: valuable therapeutic implications? J Neurol Neurosurg Psychiatry. 2007;78:1152–3.

Mantione M, van de Brink W, Schuurman PR, Denys D. Smoking cessation and weight loss after chronic deep brain stimulation of the nucleus accumbens: therapeutic and research implications: case report. Neurosurgery. 2010;66:E218.

Levy D, Shabat-Simon M, Shalev U, Barnea-Ygael N, Cooper A, Zangen A. Repeated electrical stimulation of reward-related brain regions affects cocaine but not “natural” reinforcement. J Neurosci. 2007;27:14179–89.

Liu H-Y, Jin J, Tang JS, Sun WX, Jia H, Yang XP, et al. Chronic deep brain stimulation in the rat nucleus accumbens and its effect on morphine reinforcement. Addict Biol. 2008;13:40–46.

Vassoler FM, Schmidt HD, Gerard ME, Famous KR, Ciraulo DA, Kornetsky C, et al. Deep brain stimulation of the nucleus accumbens shell attenuates cocaine priming-induced reinstatement of drug seeking in rats. J Neurosci. 2008;28:8735–9.

Batra V, Tran TL, Caputo J, Guerin GF, Goeders NE, Wilden J. Intermittent bilateral deep brain stimulation of the nucleus accumbens shell reduces intravenous methamphetamine intake and seeking in Wistar rats. J Neurosurg. 2017;126:1339–50.

Heinze H-J, et al. Counteracting incentive sensitization in severe alcohol dependence using deep brain stimulation of the nucleus accumbens: clinical and basic science aspects. Front Hum Neurosci. 2009;3:22.

Kuhn J, Gründler TO, Bauer R, Huff W, Fischer AG, Lenartz D, et al. Successful deep brain stimulation of the nucleus accumbens in severe alcohol dependence is associated with changed performance monitoring. Addict Biol. 2011;16:620–3.

Kuhn J, Möller M, Treppmann JF, Bartsch C, Lenartz D, Gruendler TO, et al. Deep brain stimulation of the nucleus accumbens and its usefulness in severe opioid addiction. Mol Psychiatry. 2014;19:145–6.

Valencia-Alfonso C-E, Luigjes J, Smolders R, Cohen MX, Levar N, Mazaheri A, et al. Effective deep brain stimulation in heroin addiction: a case report with complementary intracranial electroencephalogram. Biol Psychiatry. 2012;71:e35–37.

Voges J, Müller U, Bogerts B, Münte T, Heinze H-J. Deep brain stimulation surgery for alcohol addiction. World Neurosurg. 2013;80:S28.e21–31.

Wang TR, Moosa S, Dallapiazza RF, Elias WJ, Lynch WJ. Deep brain stimulation for the treatment of drug addiction. Neurosurg Focus. 2018;45:E11.

Sanna A, Fattore L, Badas P, Corona G, Diana M. The hypodopaminergic state ten years after: transcranial magnetic stimulation as a tool to test the dopamine hypothesis of drug addiction. Curr Opin Pharmacol. 2021;56:61–67.

Verrico CD, Haile CN, Newton TF, Kosten TR, Garza RDL. Pharmacotherapeutics for substance-use disorders: a focus on dopaminergic medications. Expert Opin Investig Drugs. 2013;22:1549–68.

Amiaz R, Levy D, Vainiger D, Grunhaus L, Zangen A. Repeated high-frequency transcranial magnetic stimulation over the dorsolateral prefrontal cortex reduces cigarette craving and consumption. Addiction. 2009;104:653–60.

Pettorruso M, Di Giuda D, Martinotti G, Cocciolillo F, De Risio L, Montemitro C, et al. Dopaminergic and clinical correlates of high-frequency repetitive transcranial magnetic stimulation in gambling addiction: a SPECT case study. Addict Behav. 2019;93:246–9.

Cho SS, Strafella AP. rTMS of the left dorsolateral prefrontal cortex modulates dopamine release in the ipsilateral anterior cingulate cortex and orbitofrontal cortex. PLoS ONE. 2009;4:e6725.

Strafella AP, Paus T, Fraraccio M, Dagher A. Striatal dopamine release induced by repetitive transcranial magnetic stimulation of the human motor cortex. Brain. 2003;126:2609–15.

Ekhtiari H, Tavakoli H, Addolorato G, Baeken C, Bonci A, Campanella S, et al. Transcranial electrical and magnetic stimulation (tES and TMS) for addiction medicine: a consensus paper on the present state of the science and the road ahead. Neurosci Biobehav Rev. 2019;104:118–40.

Wu X-Q, Zan GY, Ju YY, Chen TZ, Guo LB, Jiao DL, et al. Low-frequency repetitive transcranial magnetic stimulation inhibits the development of methamphetamine-induced conditioned place preference. Behav Brain Res. 2018;353:129–36.

Dackis CA, Kampman KM, Lynch KG, Plebani JG, Pettinati HM, Sparkman T, et al. A double-blind, placebo-controlled trial of modafinil for cocaine dependence. J Subst Abus Treat. 2012;43:303–12.

Shearer J, Darke S, Rodgers C, Slade T, van Beek I, Lewis J, et al. A double-blind, placebo-controlled trial of modafinil (200 mg/day) for methamphetamine dependence. Addiction. 2009;104:224–33.

Chiamulera C, Piva A, Abraham WC. Glutamate receptors and metaplasticity in addiction. Curr Opin Pharmacol. 2021;56:39–45.

Le Moal M, Koob GF. Drug addiction: pathways to the disease and pathophysiological perspectives. Eur Neuropsychopharmacol. 2007;17:377–93.

Luigjes J, Segrave R, de Joode N, Figee M, Denys D. Efficacy of invasive and non-invasive brain modulation interventions for addiction. Neuropsychol Rev. 2019;29:116–38.

Jordi E, Heiman M, Marion-Poll L, Guermonprez P, Cheng SK, Nairn AC, Greengard P, Girault J-A. Proceedings of the National Academy of Sciences . 2013;110:9511-16. https://doi.org/10.1073/pnas.1307116110 .

Wang H, Wang L, Erdjument-Bromage H, Vidal M, Tempst P, Jones RS, et al. Role of histone H2A ubiquitination in Polycomb silencing. Nature. 2004;431:873–8.

Download references

Acknowledgements

We thank Michele Zoli, Romain Icick and Daniel Rial for helpful comments and corrections on the manuscript. Julian Cheron is supported by a fellowship of the FRS-FNRS (Belgium). Alban de Kerchove d´Exaerde is a Research Director of the FRS-FNRS. FRS-FNRS (Belgium). Fondation Simone et Pierre Clerdent, Fondation ULB, supported this study.

Author information

Authors and affiliations.

Laboratory of Neurophysiology, ULB Neuroscience Institute, Université Libre de Bruxelles (ULB), Brussels, B-1070, Belgium

Julian Cheron & Alban de Kerchove d’Exaerde

You can also search for this author in PubMed   Google Scholar

Contributions

JC and AKE wrote the paper and AKE supervised all the work.

Corresponding author

Correspondence to Alban de Kerchove d’Exaerde .

Ethics declarations

Competing interests.

The authors declare no competing interests.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/ .

Reprints and permissions

About this article

Cite this article.

Cheron, J., Kerchove d’Exaerde, A.d. Drug addiction: from bench to bedside. Transl Psychiatry 11 , 424 (2021). https://doi.org/10.1038/s41398-021-01542-0

Download citation

Received : 05 April 2021

Revised : 14 July 2021

Accepted : 23 July 2021

Published : 12 August 2021

DOI : https://doi.org/10.1038/s41398-021-01542-0

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

This article is cited by

Snp-based and haplotype-based genome-wide association on drug dependence in han chinese.

  • Yangyang Liu

BMC Genomics (2024)

USP7/Maged1-mediated H2A monoubiquitination in the paraventricular thalamus: an epigenetic mechanism involved in cocaine use disorder

  • Julian Cheron
  • Leonardo Beccari
  • Alban de Kerchove d’Exaerde

Nature Communications (2023)

Methylation and expression quantitative trait locus rs6296 in the HTR1B gene is associated with susceptibility to opioid use disorder

  • Jianbo Zhang

Psychopharmacology (2022)

Quick links

  • Explore articles by subject
  • Guide to authors
  • Editorial policies

drug addiction research topics

U.S. flag

An official website of the United States government

The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings

Preview improvements coming to the PMC website in October 2024. Learn More or Try it out now .

  • Advanced Search
  • Journal List
  • HHS Author Manuscripts

Logo of nihpa

RANKING EVIDENCE IN SUBSTANCE USE AND ADDICTION

Hudson reddon.

1. British Columbia Centre on Substance Use, 1045 Howe Street, Vancouver, BC V6Z 2A9, Canada

2. CIHR Canadian HIV Trials Network, 588-1081 Burrard Street, Vancouver, BC V6B 3E6, Canada

Thomas Kerr

3. Department of Medicine, University of British Columbia, St. Paul’s Hospital, 1081 Burrard St, Vancouver, BC V6Z 1Y6, Canada

Hudson Reddon: Writing-Original draft, Writing-Review and Editing Thomas Kerr: Writing-Review and Editing M-J Milloy: Writing-Review and Editing

Evidence-based medicine has consistently prized the epistemological value of randomized-controlled trials (RCTs) owing to their methodological advantages over alternative designs such as observational studies. However, there are limitations to RCTs that hinder their ability to study chronic and dynamic conditions such as substance use and addiction. For these conditions, observational studies may provide superior evidence based on methodological and practical strengths. Assuming epistemic superiority of RCTs has led to an inappropriate devaluation of other study designs and the findings they support, including support for harm reduction services, especially needle exchange programs and supervised injection facilities. The value offered by observational studies should be reflected in evidence-based medicine by allowing more flexibility in evidence hierarchies that presume methodological superiority of RCTs. Despite the popularity of evidence ranking systems and hierarchies, nothing should replace critical appraisal of study methodology and examining the suitability of applying a given study design to a specific research question.

Long-regarded the gold standard of medical evidence, randomized-controlled trials (RCTs) have been given the paramount role in evaluating the safety and efficacy of new interventions to improve human health and wellbeing. Several ranking systems, including evidence hierarchies and the GRADE framework, consistently bestow superiority to the RCT and place limitations on the value that can be assigned to observational studies ( Guyatt, Oxman, Vist, et al., 2008 ; Rawlins, 2008 ; Schunemann, Fretheim, & Oxman, 2006 ). However, the limitations of RCTs are seldom acknowledged, nor is the fact that observational designs are often better suited to characterize certain health conditions, in particular, chronic diseases. Assuming epistemic superiority of RCTs has led to an inappropriate devaluation of other study designs and the findings they support. This trend is particularly important for fields that are less ethically or scientifically well-suited to RCTs, such as substance use and addiction.

Critique of current evidence ranking

The methodological strength from which RCTs draw their scientific credibility is the random allocation of the intervention among trial participants. Other primary strengths include, but are not limited to, blinding, the use of a control group and applying an intervention. Randomization is presumed to remove any between-group differences in prognostic factors associated with the development of the study outcome ( Sackett, 1996 ; Sackett, Strauss, Richardson, Rosenberg, & Haynes, 2000 ). Observational studies are able to control for known confounders, yet RCTs are the only design to address the distribution of unknown confounders through randomization. However, randomization is not a guarantee that known or unknown confounders will be balanced between groups at the outset of a given study. The distribution of covariates between the intervention and control group after randomization is typically assessed by the trialists, and if uneven distributions are observed the randomization is deemed to have failed and is repeated until balanced groups are produced. Most importantly, it is also possible that imbalances in unknown confounders exist post-randomization, which cannot be detected or corrected by study investigators. Therefore, the extent to which RCTs can remove baseline imbalances between study arms is limited, to some degree, by the existing knowledge of the disease under study (i.e., the number of known confounders that can be assessed by the study investigators post-randomization). Imbalances in confounders are also an issue for observational studies. While randomization is a more effective strategy to balance these factors than approaches used in observational studies (e.g., propensity scores), randomization does not guarantee that prognostic balance is achieved ( Austin, 2011 ; Han, Enas, & McEntegart, 2009 ; Worrall, 2010 ).

The ability to establish external validity and extrapolate a trial’s findings beyond the study protocol is a second issue—one particularly germane to RCTs in substance use and addiction. With the objective of restricting any change in study outcome to the treatment intervention, RCTs typically seek to recruit a select and specific group of participants undergoing a highly structured intervention for a brief period with limited follow-up ( Rawlins, 2008 ; Worrall, 2010 ). Patients living with comorbidities or receiving multiple medications tend to be excluded in order to minimize the heterogeneity among study participants and minimize the risk of randomizing an intervention to vulnerable patients. Among people who use illicit drugs and are living with substance use disorders, minimizing heterogeneity in the study sample is challenging due to the high prevalence of comorbid health conditions and engagement with diverse health and social services. For example, current estimates among people who inject drugs report that upwards of 50% have a lifetime diagnosis of depression, 50% are living with hepatitis C virus infection, 13% are living with HIV and the medications/treatments prescribed to these patients are highly personalized ( Conner, Pinquart, & Duberstein, 2008 ; Kessler et al., 1994 ; Lengauer, Pfeifer, & Kaiser, 2014 ; United Nations Office on Drugs and Crime, 2017 ). Notwithstanding the ethical challenges of placing restrictions on inclusion and exclusion criteria, identifying a homogeneous group of participants based on these complex comorbidities and co-interventions is often practically untenable. Administering a structured intervention to this population is further complicated by high rates of marginalization (e.g., homelessness, social instability, stigmatization) and criminalization ( Marshall et al., 2016 ; Stone et al., 2018 ). In consequence, the average treatment effect calculated from an RCT is only as valuable as the sample from which it was estimated ( Dahabreh, 2018 ; Deaton & Cartwright, 2018b ; Ioannidis, 2018 ). The superior internal validity of an RCT does not translate to invariance in the treatment effect across contexts if the sample is not representative of all the patients to which the study could be applied ( Deaton & Cartwright, 2018a , 2018b ). Nevertheless, the results of RCTs are often extrapolated generously despite the limitations imposed by selection criteria and artificial environments that do not reflect real-world applications of the target intervention ( Deaton & Cartwright, 2018a , 2018b ). As an alternative, some authors advocate for strategies such as propensity scores, instrumental variables and matching to strengthen the methodology of observational studies ( Concato & Horwitz, 2018 ; Deaton & Cartwright, 2018b ). Empirical comparisons between RCTs and well-designed observational studies have found similar summary measures of effect size with neither design producing a consistently greater effect ( Anglemyer, Horvath, & Bero, 2014 ; Benson & Hartz, 2000 ; Concato, Shah, & Horwitz, 2000 ; Sterne et al., 2002 ). Moreover, many treatments that continue to be used in clinical and non-clinical settings have been evaluated through observational methods have been found to be both safe and effective ( Concato & Horwitz, 2018 ; Deaton & Cartwright, 2018b ; Tsimberidou, Braiteh, Stewart, & Kurzrock, 2009 ). In these cases, conducting an RCT on the basis that the existing evidence is observational is unnecessary based on the long-term record of safety and effectiveness and RCTs may expose patients to unnecessary risk if patients are denied treatment shown to be beneficial ( Deaton & Cartwright, 2018b ; Vandenbroucke, 2008 ).

Some authors have also questioned the value of RCTs and evidence hierarchies for identifying unintended or adverse effects of new treatment interventions ( Osimani, 2013 ; Vandenbroucke, 2008 ). As the unintended or possibly adverse effects of novel treatments are typically unknown, the treatment allocation is masked with respect to the unintended effects even if the investigator is aware of who is receiving the treatment ( Vandenbroucke, 2004 ). These authors argue that this achieves the same separation between intervention allocation and prognosis that is accomplished through blinding ( Osimani, 2013 ). As a result, observational studies examining adverse events will not be as vulnerable to confounding and selection bias as observational studies evaluating intervention efficacy ( Vandenbroucke, 2004 , 2006 , 2008 ; Vandenbroucke & Psaty, 2008 ). This view is supported by empirical evidence that shows no systematic difference in risk estimation for adverse events between RCTs and observational studies ( Benson & Hartz, 2000 ; Concato et al., 2000 ; Papanikolaou, Christidi, & Ioannidis, 2006 ). A second critique is that the strict inclusion criteria of RCTs may exclude vulnerable participants with complex comorbidities at an increased risk of experiencing adverse events, which applies to many people who use drugs ( Conner et al., 2008 ; Rawlins, 2008 ). In these circumstances case reports or case series may be the most sensitive or only tool available to identify side effects ( Glasziou, Chalmers, Rawlins, & McCulloch, 2007 ; Stricker & Psaty, 2004 ). Based on these arguments, it has been suggested that the evidence hierarchy should be inverted when the objective is to identify unknown adverse events ( Vandenbroucke, 2008 ).

Although RCTs excel at investigating the safety and efficacy of novel pharmacological formulations, issues arise in testing harm reduction-based interventions to benefit people who inject drugs. The risk environments in which needle exchange programs (NEP) and supervised injection facilities (SIF) are implemented are highly variable between settings. For example, the criminalization of illicit drug use in North America has adjusted as policies legalizing and regulating non-medical cannabis continue to expand in the context of an opioid overdose crisis sparked by the contamination of the illicit drug supply. In these circumstances, long-term observational studies in real world or natural settings may be better suited to evaluate the evolving impacts of changes in substance use policy and criminalization than RCTs. While RCTs are not feasible or ethical in this situation, observational studies are advantageous in that they can often include more follow-up time than RCTs, which is needed to better evaluate the long-term effects of structural factors such as policy change and criminalization on the chronic and dynamic nature of substance use and addiction ( Kelly, Greene, & Bergman, 2018 ; Worrall, 2010 ). The utility of real world data has sparked calls for the integration of alternative data sources such as electronic health records and patient registries to study patient groups who may have been excluded from RCTs (e.g., elderly people living with comorbidities) ( Nabhan, Klink, & Prasad, 2019 ). Other alternatives that have been applied to evaluating substance use include pragmatic trials that blend the traits of observational studies with traditional RCTs ( Coulton et al., 2017 ; Henderson et al., 2017 ). Alternative trial designs including stepped wedge and crossover trials have been effectively applied to study diseases requiring extended follow-up evaluation ( Chotard et al., 1992 ; Hemming, Haines, Chilton, Girling, & Lilford, 2015 ). These designs have been used in diverse areas, including HIV and social policy, although they are challenging when the intervention is believed to be effective and receipt should not be denied or delayed, or there are complex contextual and patient factors that moderate the impact of the intervention on the study outcome ( Bonell et al., 2011 ; Brown & Lilford, 2006 ). Unfortunately, these challenges are common among interventions for people who use drugs. Blinding and providing a placebo are often not possible for these interventions and extrapolating the effects of the intervention beyond the study protocol must be done with caution given that substance use and addiction are chronic and relapsing conditions ( Kelly et al., 2018 ; Kelly, Greene, Bergman, White, & Hoeppner, 2019 ).

Perhaps the most significant unacknowledged weakness of RCTs is their typically limited periods for the intervention and follow-up. Due to their substantial expense, funding for post-study follow-up is typically limited to six to 24 months, which is often insufficient for chronic diseases ( Bluhm, 2010 ; Vlieland, 2002 ) such as substance use disorders, characterized as chronically relapsing conditions with recurring transitions between substance use, abstinence, treatment engagement and incarceration ( Dennis & Scott, 2007 ; Kelly et al., 2019 ). For example, the median time from first to final instance of substance use was estimated at 27 years among people admitted to publicly-funded treatment who were able to abstain for at least one year, and the average time between the first episode of substance use treatment and last instance of substance use was nine years ( Dennis, Scott, Funk, & Foss, 2005 ). A recent analyses from a national study of adults in recovery in the United States (N=39,809) found that an average of two to five attempts were required to resolve an alcohol or drug problem and the number of attempts varied by demographic characteristics, type of treatment and clinical history ( Kelly et al., 2019 ). African American ethnicity, previous use of treatment and mutual-help groups and psychiatric comorbidity were associated with a greater number of recovery attempts ( Kelly et al., 2019 ). Moreover, many patient-important outcomes including quality of life, happiness and self-esteem do not increase monotonically during recovery. These measures were found to decrease significantly in the first year following resolution of an alcohol or drug problem and then increase over subsequent years ( Kelly et al., 2018 ). As a result, the methodological strengths of RCTs are, to some extent, mitigated by the limitations of administering brief structured interventions to a heterogeneous population living with chronic and dynamic disorders. For conditions such as substance use and addiction, RCTs are often limited to providing a snapshot of the effectiveness of interventions in the short term and may fail to identify long-term effects among heterogeneous groups of participants living with chronic and dynamic disorders. In these circumstances, observational studies are often better suited to study interventions designed for a more representative sample of individuals who are often excluded from studies requiring randomization, and who require long-term follow to evaluate enduring changes in study outcomes ( Bluhm, 2010 ; Worrall, 2010 ). Rather than viewing observational designs as a next best option in situations where RCTs are perceived as practically or ethically unfeasible, it should be recognized that the methodological strengths of observational studies may provide the best available evidence to evaluate the course of chronic conditions and the effect of interventions to address them ( Bluhm, 2010 ; Worrall, 2010 ).

In contrast to the approach of evidence-based medicine that strives to identify the most accurate estimate of an average treatment effect, other researchers have proposed a framework that conceptualizes health interventions as “evidence-making” ( Rhodes & Lancaster, 2019 ). Rather than assuming a singular and universal effect of an intervention, the evidence-making framework posits that the variability of practice and patients produces multiple realities of the effects of an intervention and treats evidence, interventions and their effects as emergent, contingent and multiple ( Rhodes & Lancaster, 2019 ). By recognizing that the diversity of practices creates multiple realities of an intervention, evidence-making frameworks foster dialogue across diverse forms of knowledge and knowledge actors to recognize how the politics of intervention knowledge and the realities they create influence their effects across settings ( Rhodes & Lancaster, 2019 ). While it is beyond the scope of this paper to provide a detailed analysis of this framework, similar approaches have been raised by authors who advocate for a process of cumulative scientific understanding that is often challenged by the assumptions of evidence-based medicine and evidence hierarchies ( Deaton & Cartwright, 2018b ). The production of new scientific evidence should build upon and be integrated with existing knowledge to enhance collective scientific knowledge. Unfortunately, it is not uncommon for the findings from observational studies to be dismissed if more recent evidence from a randomized study contradicts the original results ( Deaton & Cartwright, 2018a , 2018b ). Some experts argue that new findings must be able to explain or be integrated with previous results as knowledge advances, even if previous results are believed to be invalid ( Deaton & Cartwright, 2018b ). Failure to integrate results from randomized and non-randomized studies undermines the responsibility of science to advance the cumulative understanding of health interventions and acknowledge the legitimate variability of intervention effects created by the diversity of practice and patients ( Concato et al., 2000 ; Rhodes & Lancaster, 2019 ).

The inappropriate devaluation of observational studies has slowed the implementation and scale-up of several harm reduction interventions for PWUD. The observational evidence supporting SIF and NEP includes many large-scale prospective cohort studies with several years of follow-up from multiple countries ( Aspinall et al., 2014 ; Potier, Laprevote, Dubois-Arber, Cottencin, & Rolland, 2014 ; Wood, Tyndall, Montaner, & Kerr, 2006 ). The advantages of being able to prospectively evaluate the health impacts of these interventions among heterogeneous samples of people who use drugs in real-world settings are unique methodological strengths that provide evidence into the long-term trajectories of this population. Existing evaluations of NEP and SIF have provided strong evidence for reducing HIV risk behaviours, overdose mortality and increasing engagement with addiction treatment services without adverse effects on broader public health and safety ( Aspinall et al., 2014 ; Marshall, Milloy, Wood, Montaner, & Kerr, 2011 ; Potier et al., 2014 ). In spite of this evidence, these services remain limited in many countries contending with the challenges of substance use harms ( Degenhardt et al., 2014 ).

Although ideological opposition to harm reduction-based interventions like NEP and SIF remains the primary barrier limiting their availability in many settings worldwide ( Nadelmann & LaSalle, 2017 ), their establishment and expansion has also been hindered by an ironic epistemic predicament regarding their scientific evaluation. As these services provide self-evident health and safety benefits to profoundly marginalized and vulnerable individuals, regulators and trialists have determined it would be unethical to evaluate them through RCTs as randomization would restrict the control group from accessing potentially life-saving interventions ( Bastos & Strathdee, 2000 ; Bluthenthal & Kral, 2010 ; Lurie, 1998 ). Yet, when observational evidence accumulates demonstrating the benefit of these interventions, the lack of RCTs is critiqued as a methodological weakness ( Bluthenthal & Kral, 2010 ). In effect, the anticipated benefit of these services limits the credibility of subsequent empirical evidence supporting these services ( Bluthenthal & Kral, 2010 ). Throughout the implementation of NEP and SIF, the use of observational methodologies to evaluate these services has often been referenced as a precaution against their expansion or a methodological limitation ( Palmateer et al., 2010 ; Wood et al., 2006 ).

In addition, the effectiveness of SIFs has been challenged based on uncertainty regarding the effect size of these services, despite the large evidence base that is nearly unanimous in support of SIF, with no SIF overdose deaths reported to date and no reported adverse effects ( Caulkins, Pardo, & Kilmer, 2019 ; May, Holloway, & Bennett, 2019 ). Criticism about effect heterogeneity relates to the EBM objective of estimating a singular treatment effect, whereas it is entirely possible, and we would argue likely, that the effect size of these interventions may vary across patient populations and contexts ( Deaton & Cartwright, 2018b ; Rhodes & Lancaster, 2019 ). Rather than this heterogeneity being perceived as a weakness of study methodology, we would argue that this variability is consistent with the evidence-making intervention framework whereby the diversity of practice and patients creates multiple realities of an intervention’s effects ( Rhodes & Lancaster, 2019 ). In fact, this variability should be expected based on the specific crisis situations in which these services are often implemented ( Caulkins et al., 2019 ). SIFs have been established in diverse countries experiencing different types of illicit substance use, patient populations, service models, local contexts, adjunct services and drug policy ( Caulkins et al., 2019 ). These situations are not well-suited to the evidence-based medicine perspective that attempts to minimize patient and intervention heterogeneity to estimate a precise and singular treatment effect ( Deaton & Cartwright, 2018b ; Rhodes & Lancaster, 2019 ). Instead, the existence of multiple contingent effects of an intervention that are adaptive is in keeping with the ontology of the evidence-making intervention framework ( Rhodes & Lancaster, 2019 ). While we acknowledge that randomizing participants to SIF participation (e.g., through community-randomized trials or wait-list studies) would be theoretically valuable, we contend that this is has become unethical and unnecessary given the evidence of benefit and absence of harm associated with these services ( Caulkins et al., 2019 ; Kennedy, Hayashi, Milloy, Wood, & Kerr, 2019 ). As previously mentioned, RCTs do not have the same benefits for assessing adverse outcomes and the risks associated with denying or delaying SIF use among vulnerable populations of PWUD through RCTs is difficult to justify ( May et al., 2019 ; Osimani, 2013 ; Vandenbroucke, 2006 ). Although RCTs are held as the gold-standard for identifying causal associations, observational and qualitative studies are able to provide valuable evidence about the causal mechanisms underlying these associations and contribute to advancing the cumulative understanding of an intervention without the need for experimental closure ( Deaton & Cartwright, 2018b ; Rhodes & Lancaster, 2019 ). The study design that provides the best evidence may therefore vary depending on the theory or research question being evaluated; this should be reflected in evidence ranking systems such as the GRADE framework and the Maryland Scientific Methods Scale that place limits on the value that can be assigned to non-randomized studies ( Alonso-Coello et al., 2016 ; Worrall, 2010 ).

There are examples where observational studies have been sufficient to demonstrate effectiveness and continue to be used clinically without the need for verification through RCTs. An analysis of oncology drugs approved by the United States Food and Drug Administration found that 31 of 68 drugs were approved without an RCT and 30 of these drugs remain fully approved ( Concato & Horwitz, 2018 ; Tsimberidou et al., 2009 ). Studies using objective endpoints were the most common among those approved and these drugs demonstrated a long-term record of efficacy and safety based on observational evidence ( Tsimberidou et al., 2009 ). There are also cases where the belief that RCTs are needed to confirm observational evidence has caused harm. The controversy surrounding the RCTs of extracorporeal membrane oxygenation (ECMO) therapy in newborns has been well described ( Bluhm, 2010 ; Truog, 1992 ). Both ECMO and the standard treatment were evolving rapidly and observational studies had already supported the benefit of ECMO ( Bartlett, 1984 ; Wung, James, Kilchevsky, & James, 1985 ). Yet, variability in the success rate of ECMO across centres prompted many researchers to call for RCTs before ECMO became an accepted therapy ( Bluhm, 2010 ). In the first trial, the one infant who received conventional therapy did not survive while all 11 who received ECMO survived ( Bartlett et al., 1985 ). In phase one of the second trial, four of the ten newborns receiving conventional therapy did not survive while all nine who received ECMO survived ( O’Rourke et al., 1989 ). In phase two, 20 patients received ECMO and 19 survived ( O’Rourke et al., 1989 ). These examples have led some experts to suggest that RCTs should not be used in cases where interventions are rapidly evolving and are potentially life saving ( Truog, 1992 ). In these cases, the interventions or their implementation may be out-dated by the time the RCT has concluded and observational studies of clinical practice (e.g., outcomes research) may provide better evidence of intervention effectiveness while avoiding some of the ethical challenges of withholding or delaying novel treatments in RCTs ( Truog, 1992 ; Worrall, 2010 ). This is particularly relevant to the situation of SIFs, which are often implemented in situations of crisis that are rapidly evolving as new challenges arise, such as the multiple waves of the opioid overdose crisis. Under these circumstances, variability in the effect should not detract from the fact the evidence is, even in the opinion of critics, “almost unanimous in its support” and is potentially life saving ( Caulkins et al., 2019 ; Kennedy et al., 2019 ; May et al., 2019 ). Performing RCTs of SIFs to ascertain a singular measure of effect should be viewed as unnecessary and unethical given the existing evidence and that observational studies may provide better information to evaluate these interventions based on the evolving context of their implementation and the potential for them to be life saving.

Future directions for addiction science and evidence ranking

An alternative observational design that has gained considerable attention, particularly for studying chronic diseases, is Mendelian randomization ( Lawlor, Harbord, Sterne, Timpson, & Davey Smith, 2008 ; Smith, 2006 ). These studies integrate genetic variants into observational epidemiology to enhance the causal inferences that can be drawn about modifiable risk factors and health outcomes ( Smith & Ebrahim, 2003 ; Youngman, Keavney, & Palmer, 2000 ). Rather than randomize participants to receive the exposure being studied, Mendelian randomization studies take advantage of the random assignment of an individual’s genotype from their parents to conduct a ‘natural’ RCT ( Davey Smith & Ebrahim, 2005 ; Hingorani & Humphries, 2005 ). If germline genetic variants for an environmental exposure (e.g., substance use) have been identified, these variants can be used as a proxy measure of the exposure in observational studies and be treated as randomly distributed ( Lawlor et al., 2008 ; Pasman et al., 2018 ). Mendelian randomization studies can also be performed in representative population samples without the need for exclusion criteria or randomizing participants as is necessary in traditional RCTs ( Lawlor et al., 2008 ; Smith & Ebrahim, 2003 ). In addition, the associations between these gene variants and health outcomes are not susceptible to reverse causality as germline genotypes are not affected by disease progression, and, if the gene variant is not pleiotropic, the risk of confounding is mitigated ( Smith & Ebrahim, 2004 ). Finally, genetic variants that predict an environmental exposure typically do so throughout the life span, a fact which minimizes regression dilution bias ( Smith & Ebrahim, 2004 ). As the cost of genome sequencing continues to decrease and more large-scale consortia are forming, integrating genetic data into observational designs is becoming increasingly feasible for studies of substance use and addiction ( Pasman et al., 2018 ; Phillips, Deverka, Hooker, & Douglas, 2018 ; Vaucher et al., 2018 ). For example, Mendelian randomization studies have been applied to investigate the longstanding debate surrounding the association between schizophrenia and cannabis use ( Gage et al., 2017 ; Pasman et al., 2018 ; Vaucher et al., 2018 ). The limitations of Mendelian randomization studies include population stratification, identifying a reliable genetic variant for the exposure of interest that does not have pleiotropic effects on the outcome and is not in linkage disequilibrium with other gene variants associated with the outcome ( Cardon & Palmer, 2003 ; Lawlor et al., 2008 ; Smith & Ebrahim, 2004 ). Additional challenges include developmental compensation, contextual influences on the exposures predicted by gene variants and non-linear associations ( Gibson & Wagner, 2000 ). Despite these limitations, Mendelian randomization studies retain the ability of traditional observational studies to study the development of chronic conditions over time with the advantage of studying gene variants for exposures that are randomly distributed ( Lawlor et al., 2008 ; Smith & Ebrahim, 2003 ). The ability to combine the strengths of observational studies while adding a component of randomization should challenge the assumed superiority of RCTs among policymakers and healthcare professionals. The evidence from these studies may also prevent the need to conduct RCTs to address certain research questions. For example, a Mendelian randomization study evaluating the effects of selenium supplementation on prostate cancer risk provided evidence of no effect, which was consistent with the largest ever prostate cancer prevention trial ( Yarmolinsky et al., 2018 ). This trial was stopped before completion based on a lack of efficacy and adverse events, at a cost of $114 million ( Yarmolinsky et al., 2018 ). As the efficacy and adverse events were predicted by this Mendelian randomization study, these designs may be an efficient and affordable means to study interventions traditionally assessed RCTs as big data resources for genetic information continue to expand ( Yarmolinsky et al., 2018 ).

A second area for improvement in substance use and addiction research is the further integration of patient-important outcomes. The underrepresentation of patient-reported outcomes has been highlighted in several study designs yet is more pronounced in RCTs ( Pardo-Hernandez & Alonso-Coello, 2017 ; Saldanha et al., 2017 ). Previous field-specific systematic reviews of RCTs have found that less than 25% include a patient-important outcome as a primary outcome ( Gaudry et al., 2017 ; Rahimi, Malhotra, Banning, & Jenkinson, 2010 ). The lack of patient-important outcomes is highly germane to substance use research and a systematic review is currently ongoing to identify patient-important outcomes to assess the effectiveness of opioid use disorder in the context of the overdose epidemic ( Dennis et al., 2015 ; Sanger et al., 2018 ). Increased recognition of the value of patient-important data is reflected in the Grading of Recommendations, Assessment, Development and Evaluation (GRADE) system, which recommends their inclusion at two different stages (the outset of study design and when ranking the importance of study outcomes) ( Guyatt, Oxman, Kunz, et al., 2008 ). Other frameworks designed to help stakeholders translate scientific evidence to healthcare decisions prioritize the inclusion of patient-important outcomes and considers the degree to which patients value each outcome ( Alonso-Coello et al., 2016 ). The Core Outcome Measures in Effectiveness Trials (COMET) initiative extends this view by promoting the selection of a standardized set of outcomes for each health condition to compare effectiveness across studies (“ The COMET Initiative,” 2010 ; Gorst et al., 2016 ; Williamson et al., 2012 ). The lack of patient-important outcomes in addiction literature, and the fact that these measures vary significantly in different stages of addiction, calls for further integration of these outcomes in future research ( Kelly et al., 2018 ; Sanger et al., 2018 ). Given the value of patient-important outcomes—and that they are prioritized in stakeholder frameworks including GRADE and COMET—further evaluation of these outcomes will be important to promote the uptake of observational evidence on substance use and addiction among policymakers and healthcare officials ( Gorst et al., 2016 ; Guyatt, Oxman, Kunz, et al., 2008 ; Pardo-Hernandez & Alonso-Coello, 2017 ; Williamson et al., 2012 ).

There are, without question, important limitations to studies with non-randomized designs. However, the value offered by observational studies should be reflected in evidence-based medicine by allowing more flexibility in evidence hierarchies that presume methodological superiority of RCTs. Observational designs may provide the best evidence to evaluate interventions to address chronic conditions such as substance use and addiction. Unfortunately, assuming epistemic superiority of RCTs has unnecessarily slowed the uptake of substance use research and harm reduction services for people who use drugs. Despite the popularity of evidence ranking systems and hierarchies, nothing should replace critical appraisal of study methodology and examining the suitability of applying a given study design to a specific clinical question.

Acknowledgments:

This work was supported by the CIHR Canadian HIV Trials Network (CTN 222). Dr. Hudson Reddon is supported by a Sponsor/CTN Postdoctoral Fellowship Award and a Michael Smith Research Trainee Award. Dr. M-J Milloy is supported in part by the United States National Institutes of Health (U01-DA021525), a New Investigator Award from CIHR and a Scholar Award from MSFHR. His institution has received an unstructured gift to support him from NG Biomed, Ltd., a private firm applying for a government license to produce cannabis. The Canopy Growth professorship in cannabis science was established through unstructured gifts to the University of British Columbia from Canopy Growth, a licensed producer of cannabis, and the Ministry of Mental Health and Addictions of the Government of British Columbia.

Competing Interests: None to declare.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

  • Alonso-Coello P, Schunemann HJ, Moberg J, Brignardello-Petersen R, Akl EA, Davoli M, … Group, G. W. (2016). GRADE Evidence to Decision (EtD) frameworks: a systematic and transparent approach to making well informed healthcare choices. 1: Introduction . BMJ , 353 , i2016. doi: 10.1136/bmj.i2016 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Anglemyer A, Horvath HT, & Bero L. (2014). Healthcare outcomes assessed with observational study designs compared with those assessed in randomized trials . Cochrane Database Syst Rev ( 4 ), MR000034. doi: 10.1002/14651858.MR000034.pub2 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Aspinall EJ, Nambiar D, Goldberg DJ, Hickman M, Weir A, Van Velzen E, … Hutchinson SJ (2014). Are needle and syringe programmes associated with a reduction in HIV transmission among people who inject drugs: a systematic review and meta-analysis . Int J Epidemiol , 43 ( 1 ), 235–248. doi: 10.1093/ije/dyt243 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Austin PC (2011). An Introduction to Propensity Score Methods for Reducing the Effects of Confounding in Observational Studies . Multivariate Behav Res , 46 ( 3 ), 399–424. doi: 10.1080/00273171.2011.568786 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Bartlett RH (1984). Extracorporeal oxygenation in neonates . Hosp Pract (Off Ed) , 19 ( 4 ), 139–143, 146, 151. doi: 10.1080/21548331.1984.11702803 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Bartlett RH, Roloff DW, Cornell RG, Andrews AF, Dillon PW, & Zwischenberger JB (1985). Extracorporeal circulation in neonatal respiratory failure: a prospective randomized study . Pediatrics , 76 ( 4 ), 479–487. [ PubMed ] [ Google Scholar ]
  • Bastos FI, & Strathdee SA (2000). Evaluating effectiveness of syringe exchange programmes: current issues and future prospects . Soc Sci Med , 51 , 1771–1782. [ PubMed ] [ Google Scholar ]
  • Benson K, & Hartz AJ (2000). A comparison of observational studies and randomized, controlled trials . Am J Ophthalmol , 130 ( 5 ), 688. doi: 10.1016/s0002-9394(00)00754-6 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Bluhm R. (2010). The epistemology and ethics of chronic disease research: Further lessons from ECMO . Theory of Medical Bioethics , 31 , 107–122. doi: 10.1007/s11017-010-9139-8 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Bluthenthal R, & Kral A. (2010). Commentary on Palmateer et al. (2010): next steps in the global research agenda on syringe access for injection drug users . Addiction , 105 ( 5 ), 860–861. doi: 10.1111/j.1360-0443.2010.02942.x [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Bonell CP, Hargreaves J, Cousens S, Ross D, Hayes R, Petticrew M, & Kirkwood BR (2011). Alternatives to randomisation in the evaluation of public health interventions: design challenges and solutions . J Epidemiol Community Health , 65 ( 7 ), 582–587. doi: 10.1136/jech.2008.082602 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Brown CA, & Lilford RJ (2006). The stepped wedge trial design: a systematic review . BMC Med Res Methodol , 6 , 54. doi: 10.1186/1471-2288-6-54 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Cardon LR, & Palmer LJ (2003). Population stratification and spurious allelic association . Lancet , 361 ( 9357 ), 598–604. doi: 10.1016/S0140-6736(03)12520-2 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Caulkins JP, Pardo B, & Kilmer B. (2019). Supervised consumption sites: a nuanced assessment of the causal evidence . Addiction , 114 ( 12 ), 2109–2115. doi: 10.1111/add.14747 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Chotard J, Inskip HM, Hall AJ, Loik F, Mendy M, Whittle H, … Lowe Y. (1992). The Gambia Hepatitis Intervention Study: follow-up of a cohort of children vaccinated against hepatitis B . J Infect Dis , 166 ( 4 ), 764–768. doi: 10.1093/infdis/166.4.764 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • The COMET Initiative . (2010). [ Google Scholar ]
  • Concato J, & Horwitz RI (2018). Randomized trials and evidence in medicine: A commentary on Deaton and Cartwright . Soc Sci Med , 210 , 32–36. doi: 10.1016/j.socscimed.2018.04.010 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Concato J, Shah N, & Horwitz RI (2000). Randomized, controlled trials, observational studies, and the hierarchy of research designs . N Engl J Med , 342 ( 25 ), 1887–1892. doi: 10.1056/NEJM200006223422507 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Conner KR, Pinquart M, & Duberstein PR (2008). Meta-analysis of depression and substance use and impairment among intravenous drug users (IDUs) . Addiction , 103 ( 4 ), 524–534. doi: 10.1111/j.1360-0443.2007.02118.x [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Coulton S, Stockdale K, Marchand C, Hendrie N, Billings J, Boniface S, … Wilson E. (2017). Pragmatic randomised controlled trial to evaluate the effectiveness and cost effectiveness of a multi-component intervention to reduce substance use and risk-taking behaviour in adolescents involved in the criminal justice system: A trial protocol (RISKIT-CJS) . BMC Public Health , 17 ( 1 ), 246. doi: 10.1186/s12889-017-4170-6 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Dahabreh IJ (2018). Randomization, randomized trials, and analyses using observational data: A commentary on Deaton and Cartwright . Soc Sci Med , 210 , 41–44. doi: 10.1016/j.socscimed.2018.05.012 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Davey Smith G, & Ebrahim S. (2005). What can mendelian randomisation tell us about modifiable behavioural and environmental exposures? BMJ , 330 ( 7499 ), 1076–1079. doi: 10.1136/bmj.330.7499.1076 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Deaton A, & Cartwright N. (2018a). Reflections on Randomized Control Trials . Soc Sci Med , 210 , 86–90. doi: 10.1016/j.socscimed.2018.04.046 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Deaton A, & Cartwright N. (2018b). Understanding and misunderstanding randomized controlled trials . Soc Sci Med , 210 , 2–21. doi: 10.1016/j.socscimed.2017.12.005 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Degenhardt L, Mathers BM, Wirtz AL, Wolfe D, Kamarulzaman A, Carrieri MP, … Beyrer C. (2014). What has been achieved in HIV prevention, treatment and care for people who inject drugs, 2010–2012? A review of the six highest burden countries . International Journal of Drug Policy , 25 ( 1 ), 53–60. doi: 10.1016/j.drugpo.2013.08.004 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Dennis BB, Roshanov PS, Naji L, Bawor M, Paul J, Plater C, … Thabane L. (2015). Opioid substitution and antagonist therapy trials exclude the common addiction patient: a systematic review and analysis of eligibility criteria . Trials , 16 , 475. doi: 10.1186/s13063-015-0942-4 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Dennis M, Scott C, Funk R, & Foss M. (2005). The duration and correlates of addiction and treatment careers . J Subst Abuse Treat , 28 ( 1 ), S61–S62. [ PubMed ] [ Google Scholar ]
  • Dennis M, & Scott CK (2007). Managing addiction as a chronic condition . Addict Sci Clin Pract , 4 ( 1 ), 45–55. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Gage SH, Jones HJ, Burgess S, Bowden J, Davey Smith G, Zammit S, & Munafo MR (2017). Assessing causality in associations between cannabis use and schizophrenia risk: a two-sample Mendelian randomization study . Psychol Med , 47 ( 5 ), 971–980. doi: 10.1017/S0033291716003172 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Gaudry S, Messika J, Ricard JD, Guillo S, Pasquet B, Dubief E, … Tubach F. (2017). Patient-important outcomes in randomized controlled trials in critically ill patients: a systematic review . Ann Intensive Care , 7 ( 1 ), 28. doi: 10.1186/s13613-017-0243-z [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Gibson G, & Wagner G. (2000). Canalization in evolutionary genetics: a stabilizing theory? Bioessays , 22 ( 4 ), 372–380. doi: 10.1002/(SICI)1521-1878(200004)22:4<372::AID-BIES7>3.0.CO;2-J [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Glasziou P, Chalmers I, Rawlins M, & McCulloch P. (2007). When are randomised trials unnecessary? Picking signal from noise . BMJ , 334 ( 7589 ), 349–351. doi: 10.1136/bmj.39070.527986.68 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Gorst SL, Gargon E, Clarke M, Blazeby JM, Altman DG, & Williamson PR (2016). Choosing Important Health Outcomes for Comparative Effectiveness Research: An Updated Review and User Survey . PLoS One , 11 ( 1 ), e0146444. doi: 10.1371/journal.pone.0146444 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Guyatt GH, Oxman AD, Kunz R, Vist GE, Falck-Ytter Y, Schunemann HJ, & Group GW (2008). What is “quality of evidence” and why is it important to clinicians? BMJ , 336 ( 7651 ), 995–998. doi: 10.1136/bmj.39490.551019.BE [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Guyatt GH, Oxman AD, Vist GE, Kunz R, Falck-Ytter Y, Alonso-Coello P, … Group, G. W. (2008). GRADE: an emerging consensus on rating quality of evidence and strength of recommendations . BMJ , 336 ( 7650 ), 924–926. doi: 10.1136/bmj.39489.470347.AD [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Han B, Enas NH, & McEntegart D. (2009). Randomization by minimization for unbalanced treatment allocation . Stat Med , 28 ( 27 ), 3329–3346. doi: 10.1002/sim.3710 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Hemming K, Haines TP, Chilton PJ, Girling AJ, & Lilford RJ (2015). The stepped wedge cluster randomised trial: rationale, design, analysis, and reporting . BMJ , 350 , h391. doi: 10.1136/bmj.h391 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Henderson JL, Cheung A, Cleverley K, Chaim G, Moretti ME, de Oliveira C, … Szatmari P. (2017). Integrated collaborative care teams to enhance service delivery to youth with mental health and substance use challenges: protocol for a pragmatic randomised controlled trial . BMJ Open , 7 ( 2 ), e014080. doi: 10.1136/bmjopen-2016-014080 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Hingorani A, & Humphries S. (2005). Nature’s randomised trials . Lancet , 366 ( 9501 ), 1906–1908. doi: 10.1016/S0140-6736(05)67767-7 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Ioannidis JPA (2018). Randomized controlled trials: Often flawed, mostly useless, clearly indispensable: A commentary on Deaton and Cartwright . Soc Sci Med , 210 , 53–56. doi: 10.1016/j.socscimed.2018.04.029 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Kelly J, Greene C, & Bergman B. (2018). Beyond abstinence: Changes in indices of quality of lifewith time in recovery in a nationally-representative sample of US adults . Alcohol Clin Exp Res , 42 ( 4 ), 770–780. doi: 10.1111/acer.13604 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Kelly JF, Greene MC, Bergman BG, White WL, & Hoeppner BB (2019). How Many Recovery Attempts Does it Take to Successfully Resolve an Alcohol or Drug Problem? Estimates and Correlates From a National Study of Recovering U.S. Adults . Alcohol Clin Exp Res , 43 ( 7 ), 1533–1544. doi: 10.1111/acer.14067 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Kennedy MC, Hayashi K, Milloy MJ, Wood E, & Kerr T. (2019). Supervised injection facility use and all-cause mortality among people who inject drugs in Vancouver, Canada: A cohort study . PLoS Med , 16 ( 11 ), e1002964. doi: 10.1371/journal.pmed.1002964 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Kessler RC, McGonagle KA, Zhao S, Nelson CB, Hughes M, Eshleman S, … Kendler KS (1994). Lifetime and 12-month prevalence of DSM-III-R psychiatric disorders in the United States. Results from the National Comorbidity Survey . Arch Gen Psychiatry , 51 ( 1 ), 8–19. doi: 10.1001/archpsyc.1994.03950010008002 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Lawlor DA, Harbord RM, Sterne JA, Timpson N, & Davey Smith G. (2008). Mendelian randomization: using genes as instruments for making causal inferences in epidemiology . Stat Med , 27 ( 8 ), 1133–1163. doi: 10.1002/sim.3034 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Lengauer T, Pfeifer N, & Kaiser R. (2014). Personalized HIV therapy to control drug resistance . Drug Discov Today Technol , 11 , 57–64. doi: 10.1016/j.ddtec.2014.02.004 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Lurie P. (1998). Re: ‘Invited commentary: le mystere de Montreal’ . Am J Epidemiol , 148 , 715–716. [ PubMed ] [ Google Scholar ]
  • Marshall BD, Elston B, Dobrer S, Parashar S, Hogg RS, Montaner JS, … Milloy MJ (2016). The population impact of eliminating homelessness on HIV viral suppression among people who use drugs . AIDS , 30 ( 6 ), 933–942. doi: 10.1097/qad.0000000000000990 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Marshall BD, Milloy MJ, Wood E, Montaner JS, & Kerr T. (2011). Reduction in overdose mortality after the opening of North America’s first medically supervised safer injecting facility: a retrospective population-based study . Lancet , 377 ( 9775 ), 1429–1437. doi: 10.1016/S0140-6736(10)62353-7 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • May T, Bennett T, & Holloway K. (2018). RETRACTED: The impact of medically supervised injection centres on drug-related harms: a meta-analysis . Int J Drug Policy , 59 , 98–107. doi: 10.1016/j.drugpo.2018.06.018 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • May T, Holloway K, & Bennett T. (2019). The need to broaden and strengthen the evidence base for supervised consumption sites . Addiction , 114 ( 12 ), 2117–2118. doi: 10.1111/add.14789 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Nabhan C, Klink A, & Prasad V. (2019). Real-world Evidence-What Does It Really Mean? JAMA Oncol , 5 ( 6 ), 781–783. doi: 10.1001/jamaoncol.2019.0450 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Nadelmann E, & LaSalle L. (2017). Two steps forward, one step back: current harm reduction policy and politics in the United States . Harm Reduct J , 14 ( 1 ), 37. doi: 10.1186/s12954-017-0157-y [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • O’Rourke PP, Crone RK, Vacanti JP, Ware JH, Lillehei CW, Parad RB, & Epstein MF (1989). Extracorporeal membrane oxygenation and conventional medical therapy in neonates with persistent pulmonary hypertension of the newborn: a prospective randomized study . Pediatrics , 84 ( 6 ), 957–963. [ PubMed ] [ Google Scholar ]
  • Osimani B. (2013). Until RCT proven? On the asymmetry of evidence requirements for risk assessment . J Eval Clin Pract , 19 ( 3 ), 454–462. doi: 10.1111/jep.12039 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Palmateer N, Kimber J, Hickman M, Hutchinson S, Rhodes T, & Goldberg D. (2010). Evidence for the effectiveness of sterile injecting equipment provision in preventing hepatitis C and human immunodeficiency virus transmission among injecting drug users: a review of reviews . Addiction , 105 ( 5 ), 844–859. doi: 10.1111/j.1360-0443.2009.02888.x [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Papanikolaou PN, Christidi GD, & Ioannidis JP (2006). Comparison of evidence on harms of medical interventions in randomized and nonrandomized studies . CMAJ , 174 ( 5 ), 635–641. doi: 10.1503/cmaj.050873 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Pardo-Hernandez H, & Alonso-Coello P. (2017). Patient-important outcomes in decision-making: a point of no return . J Clin Epidemiol , 88 , 4–6. doi: 10.1016/j.jclinepi.2017.05.014 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Pasman JA, Verweij KJH, Gerring Z, Stringer S, Sanchez-Roige S, Treur JL, … Vink JM (2018). GWAS of lifetime cannabis use reveals new risk loci, genetic overlap with psychiatric traits, and a causal influence of schizophrenia . Nat Neurosci , 21 ( 9 ), 1161–1170. doi: 10.1038/s41593-018-0206-1 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Phillips KA, Deverka PA, Hooker GW, & Douglas MP (2018). Genetic Test Availability And Spending: Where Are We Now? Where Are We Going? Health Aff (Millwood) , 37 ( 5 ), 710–716. doi: 10.1377/hlthaff.2017.1427 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Potier C, Laprevote V, Dubois-Arber F, Cottencin O, & Rolland B. (2014). Supervised injection services: what has been demonstrated? A systematic literature review . Drug Alcohol Depend , 145 , 48–68. doi: 10.1016/j.drugalcdep.2014.10.012 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Rahimi K, Malhotra A, Banning AP, & Jenkinson C. (2010). Outcome selection and role of patient reported outcomes in contemporary cardiovascular trials: systematic review . BMJ , 341 , c5707. doi: 10.1136/bmj.c5707 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Rawlins MD (2008). De Testimonio: On the Evidence for Decisions about the use of Therapeutic Interventions The Harveian Oration . London: Royal College of Physicians. [ Google Scholar ]
  • Rhodes T, & Lancaster K. (2019). Evidence-making interventions in health: A conceptual framing . Soc Sci Med , 238 , 112488. doi: 10.1016/j.socscimed.2019.112488 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • ackett DL (1996). Evidence based medicine. What it is and what it isn’t . British Medical Journal , 312 , 71–72. [ Google Scholar ]
  • Sackett DL, Strauss SE, Richardson WS, Rosenberg W, & Haynes RB (2000). Evidence Based Medicine. How to Practice and Teach EBM (2 ed.). Edinburgh and London: Churchill Livingston. [ Google Scholar ]
  • Saldanha IJ, Li T, Yang C, Owczarzak J, Williamson PR, & Dickersin K. (2017). Clinical trials and systematic reviews addressing similar interventions for the same condition do not consider similar outcomes to be important: a case study in HIV/AIDS . J Clin Epidemiol , 84 , 85–94. doi: 10.1016/j.jclinepi.2017.02.005 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Sanger N, Shahid H, Dennis BB, Hudson J, Marsh D, Sanger S, … Samaan Z. (2018). Identifying patient-important outcomes in medication-assisted treatment for opioid use disorder patients: a systematic review protocol . BMJ Open , 8 ( 12 ), e025059. doi: 10.1136/bmjopen-2018-025059 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Schunemann HJ, Fretheim A, & Oxman AD (2006). Improving the use of research evidence in guideline development: 9. Grading evidence and recommendations . Health Res Policy Syst , 4 ( 21 ). [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Smith GD (2006). Capitalising on Mendelian randomisation to assess the effects of treatment . James Lind Library . [ Google Scholar ]
  • Smith GD, & Ebrahim S. (2003). ‘Mendelian randomization’: can genetic epidemiology contribute to understanding environmental determinants of disease? Int J Epidemiol , 32 ( 1 ), 1–22. doi: 10.1093/ije/dyg070 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Smith GD, & Ebrahim S. (2004). Mendelian randomization: prospects, potentials, and limitations . Int J Epidemiol , 33 ( 1 ), 30–42. doi: 10.1093/ije/dyh132 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Sterne JA, Juni P, Schulz KF, Altman DG, Bartlett C, & Egger M. (2002). Statistical methods for assessing the influence of study characteristics on treatment effects in ‘meta-epidemiological’ research . Stat Med , 21 ( 11 ), 1513–1524. doi: 10.1002/sim.1184 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Stone J, Fraser H, Lim AG, Walker JG, Ward Z, MacGregor L, … Vickerman P. (2018). Incarceration history and risk of HIV and hepatitis C virus acquisition among people who inject drugs: a systematic review and meta-analysis . Lancet Infect Dis , 18 ( 12 ), 1397–1409. doi: 10.1016/S1473-3099(18)30469-9 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Stricker BH, & Psaty BM (2004). Detection, verification, and quantification of adverse drug reactions . BMJ , 329 ( 7456 ), 44–47. doi: 10.1136/bmj.329.7456.44 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Truog RD (1992). Randomized controlled trials: lessons from ECMO . Clin Res , 40 ( 3 ), 519–527. [ PubMed ] [ Google Scholar ]
  • Tsimberidou AM, Braiteh F, Stewart DJ, & Kurzrock R. (2009). Ultimate fate of oncology drugs approved by the us food and drug administration without a randomized Trial . J Clin Oncol , 27 ( 36 ), 6243–6250. doi: 10.1200/JCO.2009.23.6018 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • United Nations Office on Drugs and Crime. (2017). World Drug Report, United Nations publication (ISBN: 978–92-1–148291-1, eISBN:978–92-1–060623-3, Sales No. E.17.XI.6 ). [ Google Scholar ]
  • Vandenbroucke JP (2004). When are observational studies as credible as randomised trials? Lancet , 363 ( 9422 ), 1728–1731. doi: 10.1016/S0140-6736(04)16261-2 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Vandenbroucke JP (2006). What is the best evidence for determining harms of medical treatment? CMAJ , 174 ( 5 ), 645–646. doi: 10.1503/cmaj.051484 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Vandenbroucke JP (2008). Observational research, randomised trials, and two views of medical science . PLoS Med , 5 ( 3 ), e67. doi: 10.1371/journal.pmed.0050067 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Vandenbroucke JP, & Psaty BM (2008). Benefits and risks of drug treatments: how to combine the best evidence on benefits with the best data about adverse effects . JAMA , 300 ( 20 ), 2417–2419. doi: 10.1001/jama.2008.723 [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Vaucher J, Keating BJ, Lasserre AM, Gan W, Lyall DM, Ward J, … Holmes MV (2018). Cannabis use and risk of schizophrenia: a Mendelian randomization study . Mol Psychiatry , 23 ( 5 ), 1287–1292. doi: 10.1038/mp.2016.252 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Vlieland T. (2002). Managing chronic disease: Evidence-based medicine or patient centred medicine? Health Care Analysis , 10 ( 3 ), 289–298. [ PubMed ] [ Google Scholar ]
  • Williamson PR, Altman DG, Blazeby JM, Clarke M, Devane D, Gargon E, & Tugwell P. (2012). Developing core outcome sets for clinical trials: issues to consider . Trials , 13 , 132. doi: 10.1186/1745-6215-13-132 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Wood E, Tyndall MW, Montaner JS, & Kerr T. (2006). Summary of findings from the evaluation of a pilot medically supervised safer injecting facility . CMAJ , 175 ( 11 ), 1399–1404. doi: 10.1503/cmaj.060863 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Worrall J. (2010). Evidence: philosophy of science meets medicine . J Eval Clin Pract , 16 ( 2 ), 356–362. doi: 10.1111/j.1365-2753.2010.01400.x [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Wung JT, James LS, Kilchevsky E, & James E. (1985). Management of infants with severe respiratory failure and persistence of the fetal circulation, without hyperventilation . Pediatrics , 76 ( 4 ), 488–494. [ PubMed ] [ Google Scholar ]
  • Yarmolinsky J, Bonilla C, Haycock PC, Langdon RJQ, Lotta LA, Langenberg C, … Martin RM (2018). Circulating Selenium and Prostate Cancer Risk: A Mendelian Randomization Analysis . J Natl Cancer Inst , 110 ( 9 ), 1035–1038. doi: 10.1093/jnci/djy081 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Youngman LD, Keavney BD, & Palmer A. (2000). Plasma fibrinogen and fibrinogen genotypes in 4685 cases of myocardial infarction and in 6002 controls: test of causality by ‘Mendelian randomization’ . Circulation , 102 ( Suppl. II ), 31–32. [ Google Scholar ]

Logo

  • Signs of Addiction

Addiction Research

Discover the latest in addiction research, from the neuroscience of substance use disorders to evidence-based treatment practices. reports, updates, case studies and white papers are available to you at hazelden betty ford’s butler center for research..

Man Writing paper

Why do people become addicted to alcohol and other drugs? How effective is addiction treatment? What makes certain substances so addictive? The Butler Center for Research at the Hazelden Betty Ford Foundation investigates these and other questions and publishes its scientific findings in a variety of alcohol and drug addiction research papers and reports. Research topics include:

  • Evidence-based treatment practices
  • Addiction treatment outcomes
  • Addiction, psychiatry and the brain
  • Addictive substances such as prescription opioids and heroin
  • Substance abuse in youth/teens, older adults and other demographic groups such as health care or legal professionals

These research queries and findings are presented in the form of updates, white papers and case studies. In addition, the Butler Center for Research collaborates with the Recovery Advocacy team to study special-focus addiction research topics, summarized in monthly  Emerging Drug Trends  reports. Altogether, these studies provide the latest in addiction research for anyone interested in learning more about the neuroscience of addiction and how addiction affects individuals, families and society in general. The research also helps clinicians and health care professionals further understand, diagnose and treat drug and alcohol addiction. Learn more about each of the Butler Center's addiction research studies below.

Research Updates

Written by Butler Center for Research staff, our one-page, topic-specific summaries discuss current research on topics of interest within the drug abuse and addiction treatment field.

View our most recent updates, or view the archive at the bottom of the page.

Patient Outcomes Study Results at Hazelden Betty Ford

Trends and Patterns in Cannabis Use across Different Age Groups

Alcohol and Tobacco Harm Reduction Interventions

Harm Reduction: History and Context

Racial and Ethnic Health Disparities and Addiction

Psychedelics as Therapeutic Treatment

Sexual and Gender Minority Youth and SUDs

Health Care Professionals and Mental Health

Grief and Addiction

Helping Families Cope with Addiction

Emerging Drug Trends Report and National Surveys

Shedding New Light on America’s No. 1 Health Problem

In collaboration with the University of Maryland School of Public Health and with support from the Butler Center for Research, the Recovery Advocacy team routinely issues research reports on emerging drug trends in America. Recovery Advocacy also commissions national surveys on attitudes, behaviors and perspectives related to substance use. From binge drinking and excessive alcohol use on college campuses, to marijuana potency concerns in an age of legalized marijuana, deeper analysis and understanding of emerging drug trends allows for greater opportunities to educate, inform and prevent misuse and deaths.

Each drug trends report explores the topic at hand, documenting the prevalence of the problem, relevant demographics, prevention and treatment options available, as well as providing insight and perspectives from thought leaders throughout the Hazelden Betty Ford Foundation.

View the latest  Emerging Drug Trends  Report:

Pediatricians First Responders for Preventing Substance Use

  • Clearing Away the Confusion: Marijuana Is Not a Public Health Solution to the Opioid Crisis
  • Does Socioeconomic Advantage Lessen the Risk of Adolescent Substance Use?
  • The Collegiate Recovery Movement Is Gaining Strength
  • Considerations for Policymakers Regarding Involuntary Commitment for Substance Use Disorders
  • Widening the Lens on the Opioid Crisis
  • Concerns Rising Over High-Potency Marijuana Use
  • Beyond Binging: “High-Intensity Drinking”

View the latest  National Surveys :

  • College Administrators See Problems As More Students View Marijuana As Safe

College Parents See Serious Problems From Campus Alcohol Use

  • Youth Opioid Study: Attitudes and Usage

About Recovery Advocacy

Our mission is to provide a trusted national voice on all issues related to addiction prevention, treatment and recovery, and to facilitate conversation among those in recovery, those still suffering and society at large. We are committed to smashing stigma, shaping public policy and educating people everywhere about the problems of addiction and the promise of recovery. Learn more about recovery advocacy and how you can make a difference.

Evidence-Based Treatment Series

To help get consumers and clinicians on the same page, the Butler Center for Research has created a series of informational summaries describing:

  • Evidence-based addiction treatment modalities
  • Distinctive levels of substance use disorder treatment
  • Specialized drug and alcohol treatment programs

Each evidence-based treatment series summary includes:

  • A definition of the therapeutic approach, level of care or specialized program
  • A discussion of applicability, usage and practice
  • A description of outcomes and efficacy
  • Research citations and related resources for more information

View the latest in this series:

Motivational Interviewing

Cognitive Behavioral Therapy

Case Studies and White Papers

Written by Hazelden Betty Ford Foundation researchers and clinicians, case studies and white papers presented by the Butler Center for Research provide invaluable insight into clinical processes and complex issues related to addiction prevention, treatment and recovery. These in-depth reports examine and chronicle clinical activities, initiatives and developments as a means of informing practitioners and continually improving the quality and delivery of substance use disorder services and related resources and initiatives.

  • What does it really mean to be providing medication-assisted treatment for opioid addiction?

Adolescent Motivational Interviewing

Peer Recovery Support: Walking the Path Together

Addiction and Violence During COVID-19

The Brain Disease Model of Addiction

Healthcare Professionals and Compassion Fatigue

Moving to Trauma-Responsive Care

Virtual Intensive Outpatient Outcomes: Preliminary Findings

Driving Under the Influence of Cannabis

Vaping and E-Cigarettes

Using Telehealth for Addiction Treatment

Grandparents Raising Grandchildren

Substance Use Disorders Among Military Populations

Co-Occurring Mental Health and Substance Use Disorders

Women and Alcohol

Prescription Rates of Opioid Analgesics in Medical Treatment Settings

Applications of Positive Psychology to Substance Use Disorder

Substance Use Disorders Among Legal Professionals

Factors Impacting Early Alcohol and Drug Use Among Youths

Animal-Assisted Therapy for Substance Use Disorders

Prevalence of Adolescent Substance Misuse

Problem Drinking Behaviors Among College Students

The Importance of Recovery Management

Substance Use Factors Among LGBTQ individuals

Prescription Opioids and Dependence

Alcohol Abuse Among Law Enforcement Officers

Helping Families Cope with Substance Dependence

The Social Norms Approach to Student Substance Abuse Prevention

Drug Abuse, Dopamine and the Brain's Reward System

Women and Substance Abuse

Substance Use in the Workplace

Health Care Professionals: Addiction and Treatment

Cognitive Improvement and Alcohol Recovery

Drug Use, Misuse and Dependence Among Older Adults

Emerging Drug Trends

Does Socioeconomic Advantage Lessen the Risk of Adolescent Substance Use

The Collegiate Recovery Movement is Gaining Strength

Involuntary Commitment for Substance Use Disorders

Widening the Lens of the Opioid Crisis

Beyond Binge Drinking: High Intensity Drinking

High Potency Marijuana

National Surveys

College Administrators See Problems as More Students View Marijuana as Safe

Risky Opioid Use Among College-Age Youth

Case Studies/ White Papers

What does it really mean to be providing medication-assisted treatment for opioid addiction

Are you or a loved one struggling with alcohol or other drugs? Call today to speak confidentially with a recovery expert. Most insurance accepted.

Harnessing science, love and the wisdom of lived experience, we are a force of healing and hope ​​​​​​​for individuals, families and communities affected by substance use and mental health conditions..

accreditation badge

IMAGES

  1. 🏆 Drug addiction research topics. 13 Common Discussion Topics in

    drug addiction research topics

  2. Substance Use 101

    drug addiction research topics

  3. project report on drugs addiction

    drug addiction research topics

  4. Drug Addiction and Youth

    drug addiction research topics

  5. 🏆 Drug addiction research topics. 13 Common Discussion Topics in

    drug addiction research topics

  6. (PDF) Drug Addiction: A review of challenges and solutions

    drug addiction research topics

VIDEO

  1. Drug addiction in Iraq: Hospitals launch rehabilitation programs

  2. Health Impacts of Policy, Drug Decrim, and Service Access Panel

  3. Neurobiological Pathways to Addiction Drive Novel Treatment Interventions

  4. ALCOHOLISM AND DRUG ADDICTION

  5. Taking Notes Of Drug Addiction// #youtube #shortsfeed #studyvlog #notes @komalsingh1

  6. Battling Addiction with Research