You are on page 1of 43

 

 
Treatment-refractory substance use disorder: Focus on alcohol, opioids, and
cocaine

Michael Soyka, Jochen Mutschler

PII: S0278-5846(15)30057-9
DOI: doi: 10.1016/j.pnpbp.2015.11.003
Reference: PNP 8850

To appear in: Progress in Neuropsychopharmacology & Biological Psychiatry

Received date: 14 September 2015


Revised date: 23 October 2015
Accepted date: 11 November 2015

Please cite this article as: Soyka Michael, Mutschler Jochen, Treatment-refractory sub-
stance use disorder: Focus on alcohol, opioids, and cocaine, Progress in Neuropsychophar-
macology & Biological Psychiatry (2015), doi: 10.1016/j.pnpbp.2015.11.003

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.
ACCEPTED MANUSCRIPT

Treatment-refractory substance use disorder: Focus on alcohol, opioids, and


cocaine
Michael Soyka, MD a, b Jochen Mutschler, MD c

T
a
Department of Psychiatry and Psychotherapy
Ludwig Maximilian University

IP
Nussbaumstrasse 7
80336 Munich, Germany

R
b
Privatklinik Meiringen

SC
Postfach 612
CH – 3860 Meiringen, Switzerland

NU
c
Center for Addictive Disorders
Department of Psychiatry, Psychotherapy and Psychosomatics
Psychiatric Hospital, University of Zurich
MA
Selnaustrasse 9
8001 Zurich, Switzerland
D

Corresponding author:
Michael Soyka, MD
TE

Privatklinik Meiringen
Postfach 612
CH–3860 Meiringen
P

Switzerland
Tel: +41 33 972 82 95
CE

Fax: +41 33 972 82 91


Email: michael.soyka@privatklinik-meiringen.ch
AC

Submitted to: Progress in Neuro-Psychopharmacology & Biological Psychiatry for inclusion in


the Special Issue “Neurobiological understanding and treatment options for treatment-refractory
psychiatric disorders”

Short title: Treatment of substance use disorders


ACCEPTED MANUSCRIPT

Abstract
Substance use disorders are common, but only a small minority of patients receive adequate
treatment. Although psychosocial therapies are effective, relapse is common. This review
focusses on novel pharmacological and other treatments for patients with alcohol, opioid, or

T
cocaine use disorders who do not respond to conventional treatments.

IP
Disulfiram, acamprosate, and the opioid antagonist naltrexone have been approved for
the treatment of alcoholism. A novel, “as needed” approach is the use of the mu-opioid antagonist

R
and partial kappa agonist nalmefene to reduce alcohol consumption. Other novel pharmacological

SC
approaches include the GABA-B receptor agonist baclofen, anticonvulsants such as topiramate
and gabapentin, the partial nicotine receptor agonist varenicline, and other drugs. For opioid

NU
dependence, opioid agonist therapy with methadone or buprenorphine is the first-line treatment
option. Other options include oral or depot naltrexone, morphine sulfate, depot or implant
formulations, and heroin (diacetylmorphine) in treatment-refractory patients. To date, no
MA
pharmacological treatment has been approved for cocaine addiction; however, 3 potential
pharmacological treatments are being studied, disulfiram, methylphenidate, and modafinil.
D

Pharmacogenetic approaches may help to optimize treatment response in otherwise


treatment-refractory patients and to identify which patients are more likely to respond to
TE

treatment, and neuromodulation techniques such as repeated transcranial magnetic stimulation


and deep brain stimulation also may play a role in the treatment of substance use disorders.
P

Although no magic bullet is in sight for treatment-refractory patients, some novel


CE

medications and brain stimulation techniques have the potential to enrich treatment options at
least for some patients.
AC

Key words: alcoholism, opioid dependence, baclofen, brain stimulation, nalmefene

2
ACCEPTED MANUSCRIPT

1. Introduction
ICD-10 and DSM-IV follow a categorical approach and classify substance use disorders (SUDs)
as abuse (harmful use) or dependence. Substance abuse/harmful use is characterized by somatic
or psychiatric problems (and social problems in DSM-IV but not ICD-10). These classifications

T
define dependence by a cluster of somatic, psychological, and behavioral symptoms (APA, 2000;

IP
WHO, 1992). The recently published DSM-5 has abandoned the categorical distinction between
abuse and dependence and introduced a dimensional approach (APA, 2013). Substance-related

R
and addictive disorders are specified by 11 symptoms: 6 or more positive symptoms constitute a

SC
severe substance use disorder; 4 or 5, a moderate one; and 2 to 3, a mild one. SUDs are associated
with high psychiatric and somatic morbidity, a substantial global burden of morbidity and

NU
premature death (Gowing et al., 2015).
Numerous studies indicate that SUDs, in particular alcoholism, are common. A recent
report on global statistics of addictive behaviors (Gowing et al., 2015) states that 4.9% of the
MA
world adult population have an alcohol use disorder (7.8% of men and 1.5% of women), 22.5%
of the adult population smoke tobacco products, and 3.5% use cannabis. The use of other illegal
D

psychoactive drugs is less than 1% for each class. The prevalence estimates are 0.2% for opioid
use and 0.5% for both cocaine and amphetamines. Recent European data suggest that 1.9% of
TE

young Europeans (15-34 years old) have used cocaine at least once in the last 12 months, and 1%
of this group have used amphetamines (EMCDDA, 2015). Opioid use is reported in 0.4% of
P

adults (15-64 years old).


CE

Earlier studies estimated the prevalence of alcoholism to be 7%-10% in Europe and the
USA (Grant et al., 2004; Kessler et al., 2005; Pirkola et al., 2006; Rehm et al., 2005). Using
AC

DSM-5 criteria, the US National Epidemiologic Survey on Alcohol and Related Conditions II
reported a 12-month and lifetime prevalence for alcohol use disorders of 13.9% and 29.1%,
respectively (Grant et al., 2015). Only 19.8% of affected people had ever been treated. Globally,
prevalence estimates of alcoholism range from 0% to 16% (WHO, 2011).
Opioid dependence is a chronic relapsing disorder with a significant mortality rate
(Degenhardt et al., 2011; Degenhardt et al., 2013; Peles et al., 2010). Epidemiological studies
indicate that the worldwide prevalence of opioid use disorders is about 0.4% in individuals aged
15-64 years and that there are 15.5 million opioid-dependent people worldwide (United Nations
Office on Drugs and Crime, 2006). Epidemiological data suggest that in the European Union
prevalence rates for opioid consumption have declined in recent years (EMCDDA, 2014);
however, still about 1.3 million individuals in the EU have problematic opioid use, with a
prevalence of about 0.4%. The drugs of choice have shifted somewhat from heroin towards other

3
ACCEPTED MANUSCRIPT

opioids, including methadone, buprenorphine, and fentanyl (EMCDDA, 2014). In the USA, some
3.7 million individuals have used heroin at least once in their lives, and 750,000 to 1 million
individuals are currently heroin dependent (Kessler et al., 2012; Kleber et al., 2007). The World
Health Organization (WHO) estimates that the burden of harm from opioid use is 11.2 million

T
disability-adjusted life years (DALYs; WHO, 2004). The Global Burden of Disease study

IP
estimated that the burden of harm from opioid dependence is 9.2 million DALYs (Degenhardt et
al., 2014; Degenhardt et al., 2013). In addition, the USA in particular has an epidemic of opioid

R
prescription drug use and has recorded multiple deaths associated with an overdose of opioid pain

SC
killers, including many accidental poisonings in children (Imtiaz et al., 2014).
Cocaine abuse is becoming increasingly prevalent in western countries. Cocaine is the

NU
second most common illicit drug (after marihuana) in both the USA and in almost all western
industrial societies. The 12-month prevalence for cocaine use is 1% in Europe, and the lifetime
prevalence is 4.6% (EMCDDA, 2015).
MA
In contrast to other psychiatric disorders, “treatment-refractory” SUDs have no clear or
operationalized definition. In a recent review on heroin treatment in treatment-refractory heroin
D

addiction, Strang et al. (2015) reported studies in patients who “repeatedly failed in orthodox
treatment.” This definition may serve well for this review. Usually, treatment of SUDs has 2
TE

goals: (1) Complete and continuing abstinence, or (2) reduction of substance use (harm reduction
strategy). Agonist drug maintenance plays an important role in the latter, especially in opioid
P

dependence. For pharmacological and other reasons, agonist maintenance treatment is not
CE

suitable for all drugs of abuse. A recently published excellent and insightful comment on this
topic is provided by Darke and Farrell (2015). Since alcohol, opioid, and cocaine use plays the
AC

most important role in substance use treatment and causes significant psychiatric and somatic
complications, this narrative will focus on these types of drugs and in particular on new or
emerging treatment options. We identified relevant publications from the years 2005-2015
through a Medline/PubMed search with the terms “pharmacotherapy,” “therapy,” and “brain
stimulation.”

2. Alcoholism
2.1 Neurobiology
The neurobiological basis of alcoholism is complex and has been the subject of intensive research
in recent years (for a review see Noronha et al., 2014). In brief, the neural substrates and
neurocircuitry of alcohol dependence and other drugs of abuse include the limbic system (ventral
tegmentum and nucleus accumbens) and orbito- and prefrontal cortices. Dopamine (DA) release

4
ACCEPTED MANUSCRIPT

in the nucleus accumbens mediates reinforcing effects of drugs of psychoactive drugs (reward
processing); the prefrontal cortex is of relevance for cognitive control and the orbitofrontal cortex
for motivation (Nutt and Nestor, 2013).
Other variables mediating the vulnerability for alcohol dependence are stress or

T
sensitivity to stress and neuroendocrine function, especially the hypothalamic-pituitary-adrenal

IP
(HPA) axis, among others (Heilig and Koob, 2007; Spanagel et al., 2014a; Stephens et al.,
2014).

R
Alcohol has different molecular targets and a low affinity to many neuroreceptors. There

SC
is no specific alcohol receptor or molecular target. Neurotransmitters affected by alcohol include
inhibitory gamma-aminobutyric acid (GABA), the opioid endorphin system, glutamate, the

NU
endocannabinoid system, noradrenaline, DA, and serotonin (Koob et al., 2014; Spanagel and
Vengeliene, 2013).
There are 3 major classes of opioid receptors: mu (µ), kappa (κ), and delta (δ)
MA
(Gianoulakis, 2004). Recent research has viewed the opioidergic system as a “hedonic” system. It
is implicated in the development of alcohol use disorders (Nutt, 2014) and mediates the
D

reinforcing effects of alcohol by indirectly modulating DA release (Gianoulakis, 2004; Koob et


al., 2014), especially via the mu-opioid receptor subtype (Narita et al., 2001).
TE

Alcohol stimulates the release of the endogenous opioid receptor ligands beta-endorphin,
enkephalins, and dynorphin (Dai et al., 2005; Marinelli et al., 2005; Marinelli et al., 2006;
P

Marinelli et al., 2004). Opioid receptors on GABAergic neurons interact with dopaminergic
CE

neurons and thus mediate DA release (Koob et al., 2014). Functional neuroimaging data suggest a
negative correlation between mu-opioid receptor binding and alcohol craving (Bencherif et al.,
AC

2004; Heinz et al., 2005).

3.2 Approved medications


Beside psychosocial therapies, medications can be used to lower relapse risk to heavy drinking,
although pharmacotherapy for alcoholism is still a widely neglected area and few
pharmacotherapies have been approved for treatment of alcoholism to date (Heilig and Egli,
2006; Soyka et al., 2011a; Soyka and Rosner, 2010; Spanagel and Kiefer, 2008).

3.2.1 Disulfiram
For decades, the acetaldehyde dehydrogenase inhibitor disulfiram was the only available drug to
treat alcohol dependence. Disulfiram is described as an aversively-acting agent and induces
negative states through an unpleasant alcohol-disulfiram reaction, mediated by acetaldehyde

5
ACCEPTED MANUSCRIPT

inhibition of hepatic aldehyde dehydrogenase (Ehrenreich and Krampe, 2004; Mutschler et


al., 2011; Soyka et al., 2011a; Soyka and Lieb, 2015). However, evidence for efficacy is
limited, at least in non-supervised settings; supervised treatment with disulfiram produces high
effect sizes, making it an effective and well-established form of pharmacotherapy for relapse

T
prevention in alcohol dependence (Skinner et al., 2014). The use of disulfiram has become

IP
controversial for various reasons, including the safety profile of the drug, patient adherence to
treatment, methodological limitations of former studies, and mainly the psychologically aversive

R
nature of the treatment approach itself (Mutschler and Kiefer, 2013).

SC
3.2.2 Acamprosate

NU
In the 1990s, the putative N-methyl-D-aspartic acid (NMDA) modulator acamprosate (Maisel et
al., 2013; Rosner et al., 2010a) was introduced into clinical practice for the treatment of
alcoholism. The mechanism of action of acamprosate is not fully understood, but some data
MA
suggest that modulation of the glutamatergic NMDA receptors is of relevance (Littleton and
Zieglgansberger, 2003). Recent data indicate that the calcium part of the molecule is the only
D

active compound, but these findings need to be replicated (Spanagel et al., 2014b). The efficacy
TE

of acamprosate was hypothesized to be linked to glutamatergic hyperactivity in the CNS


(Gueorguieva et al., 2011).
Clinically, acamprosate is usually safe and well tolerated. A Cochrane analysis suggests
P

that the only frequent side effect is diarrhea (Rosner et al., 2010a). Acamprosate is given as 3
CE

tablets of 333 mg 3 times a day in patients with a bodyweight ≥ 60 kg and 2 tablets twice a day
in patients weighing ≤ 60 kg. The comparatively high number of tablets needed may limit the
AC

acceptance of acamprosate treatment. Numerous randomized controlled trials (RCTs) on


acamprosate have been performed in more than 4000 patients in total (Rosner et al., 2010a).
Furthermore, several recent meta-analyses have evaluated acamprosate (Jonas et al., 2014;
Rosner et al., 2010a), including 1 Cochrane analysis (Rosner et al., 2010a). Although there are
some important negative studies (Anton et al., 2006; Mann et al., 2013b), the overall data show
moderate but significant effects of acamprosate on abstinence rates or relapse to heavy drinking,
a finding that is supported also by a recent Japanese study (Higuchi, 2015). Studies to date
suggest that acamprosate is associated with a reduction of the risk of returning to alcohol drinking
(Jonas et al., 2014). Unfortunately, acamprosate has not found much acceptance in clinical
practice (Knudsen and Roman, 2014; Zindel and Kranzler, 2014).

3.2.3 Naltrexone

6
ACCEPTED MANUSCRIPT

The opioid antagonist naltrexone (Maisel et al., 2013) was introduced into clinical practice also in
the 1990s. Opioid antagonists have been studied extensively in alcoholism. The nonselective
opioid antagonist naltrexone is orally active and administered as a 50 mg tablet once a day.
Naltrexone is licensed and covered by health insurance in many countries. In the USA, naltrexone

T
is also available in a depot formulation (Lobmaier et al., 2011). Naltrexone reduces the
“pleasant,” rewarding effects of alcohol. Numerous studies have been performed on naltrexone

IP
(Rosner et al., 2010b), especially in combination with psychotherapy (Jarosz et al., 2013). Similar

R
to acamprosate, oral naltrexone reduces the risk of returning to drinking alcohol (Donoghue et

SC
al., 2015; Jonas et al., 2014); however, it also is not well accepted in clinical practice (Knudsen
and Roman, 2014; Zindel and Kranzler, 2014). There also are some recent conflicting data

NU
(O'Malley et al., 2015). Recently, the combination of naltrexone with the alpha adrenergic
antagonist prazosin as an daily or “as needed” treatment was studied, with some
encouraging results in the animal model (Froehlich et al., 2013; Rasmussen et al., 2015).
MA

3.3. New pharmacological developments


D

A substantial number of preclinical and clinical compounds are currently under investigation for
the treatment of alcoholism (for a review see Davies et al., 2013; Franck and Jayaram-Lindstrom,
TE

2013; Litten et al., 2014; Muller et al., 2014; Spanagel and Vengeliene, 2013; Zindel and
Kranzler, 2014), although progress is slow (Litten et al., 2014). Some medications have been
P

approved recently or are particularly useful in otherwise refractory or difficult-to-treat patients.


CE

3.3.1. Nalmefene
AC

Nalmefene was approved in the US in the 1990s for the treatment of opioid overdose. In 2012, it
became the first medication explicitly approved by the European Medicines Agency for the
reduction of alcohol intake (EMA, 2014). It is an antagonist at the mu- and delta-opioid receptor
(Emmerson et al., 1994; Niciu and Arias, 2013) and a partial agonist at the kappa receptor (Bart
et al., 2005). The functional relevance of the kappa receptor in alcoholism is somehow unclear,
but evidence suggests that it may be of relevance for motivational aspects (Walker and Koob,
2008; Walker et al., 2012; Walker et al., 2011). The oral bioavailability of nalmefene is 40%
(EMA, 2014; Ingman et al., 2005).
Neuroimaging data indicate a slow dissociation of nalmefene from the mu-opioid
receptor (Dixon et al., 1987; Ingman et al., 2005), effective binding to central opioid receptors
(Emmerson et al., 1994; Ingman et al., 2005), and a higher bioavailability (Dixon et al., 1987)
compared to naltrexone. There is no evidence for a dose-dependent association with liver toxicity

7
ACCEPTED MANUSCRIPT

(Mason et al., 1999). Preclinical findings suggest that nalmefene is effective in alcoholism (June
et al., 1998; Koob et al., 2003; Walker and Koob, 2008; Walker et al., 2011). Similar to
naltrexone, nalmefene reduces the reinforcing effects or subjective “high” feeling after alcohol
consumption (Drobes et al., 2003; 2004). To date, 6 RCTs have been published on nalmefene

T
(Anton et al., 2004; Gual et al., 2013; Karhuvaara et al., 2007; Mann et al., 2013a; Mason et al.,

IP
1994; Mason et al., 1999), subsequent to some pilot studies with limited statistical power (Mason
et al., 1994; Mason et al., 1999).

R
Anton et al. (2004) evaluated 3 doses of nalmefene (5, 20, and 40 mg) in a double-blind

SC
comparison with placebo over a 12-week treatment period (N=270). Both the nalmefene and
placebo groups showed a reduction in heavy drinking days, and no differences were found

NU
between the groups on any relevant measures. In contrast, a multicenter, placebo-controlled RCT
by Karhuvaara et al. (2007) in 403 heavy drinkers found significant effects of nalmefene
compared to placebo on the mean number of heavy drinking days. The most common adverse
MA
events associated with nalmefene were nausea, insomnia, fatigue, and dizziness, all of which are
typical for opioid antagonists.
D

Two major placebo-controlled RCTs that explicitly focused on the efficacy of


nalmefene in reducing alcohol use were conducted in Europe (Gual et al., 2013; Mann et al.,
TE

2013a). Both studies used a novel “as needed” design (i.e. the patients could decide to take the
drug on a daily basis or not) and compared 18 mg nalmefene with placebo.
P

Mann et al. (2013a) compared nalmefene with placebo over 6 months in 579 patients
CE

with alcohol dependence. The study found a significant reduction of total daily alcohol
consumption and heavy drinking days in the nalmefene group. However, the number of patients
AC

who discontinued treatment was significantly higher in verum group. Again, the main treatment-
emergent adverse events relevant for discontinuation were nausea, dizziness, fatigue, and
headache. Gual et al. (2013) evaluated as-needed use in 718 patients. Patients took study
medication on 65% of the days in the main treatment period. From baseline to month 6, alcohol
consumption decreased and heavy drinking days were reduced significantly in the nalmefene
group compared to the placebo group. In contrast to the Mann et al. (2013a) study, the incidence
of adverse events leading to drop-out was similar in both groups. A subsequent pooled analysis of
the 2 studies (van den Brink et al., 2013) showed that nalmefene was effective in patients who did
not reduce their consumption after the initial assessment.
The novel “as needed” approach used in some of the studies may be attractive for
patients who are “treatment refractory” in other treatment settings and not yet motivated or

8
ACCEPTED MANUSCRIPT

suitable for abstinence-oriented treatment. However, to date the use and clinical efficacy of
nalmefene is still somewhat controversial (Braillon, 2014; Spence, 2014; Swift, 2013).

3.3.2. Topiramate

T
Topiramate is an antiepileptic medication approved for the treatment of epilepsy (French et al.,

IP
2004; Wenzel et al., 2006). Topiramate facilitates the inhibitory action of GABA at its non-
benzodiazepine receptor and reduces the glutamate excitatory action at the α-amino-3-hydroxy-

R
5-methyl-4-isoxazolepropionic acid (AMPA) and kainate receptors (Angehagen et al., 2005;

SC
White et al., 2000). Its mechanism of action in alcoholism is not entirely clear (Guglielmo et al.,
2015), but it is assumed to decrease the reward effects of alcohol by reducing the mesolimbic

NU
cortical activity of dopamine (Johnson et al., 2004). Topiramate has a bioavailability of at least
80% and a half-life of about 19 to 23 hours. The most common adverse events include
paresthesia, anorexia, difficulty with memory or concentration, and taste perversion (Johnson and
MA
Ait-Daoud, 2010). Most side effects are mild to moderate (Johnson, 2005; 2008). Cognitive
impairment may occur (Biton et al., 2001; Johnson, 2005) and has been described also in studies
D

in patients with alcoholism (Batki et al., 2014).


A systematic review (Blodgett et al., 2014) included 7 RCTs (N = 1,125 participants)
TE

that compared topiramate to placebo for the treatment for alcohol use disorders. Overall, this
meta-analysis showed small to moderate effects of topiramate. The largest effect was found on
P

abstinence and heavy drinking. Additional studies are available, including a negative one
CE

(Likhitsathian et al., 2013). Batki et al (2014) studied the effects of topiramate in veterans with
post-traumatic stress disorder (PTSD) and alcohol use disorder; the group reported topiramate to
AC

be effective in reducing alcohol consumption and craving as well as PTSD symptom severity.
Kranzler et al. (2014a; 2014b) reported moderating effects of a single nucleotide
polymorphism (rs2832407) in GRIK1 that encodes the glutamatergic kainate GluK1 receptor
subunit. The effect on heavy drinking days was significantly greater than for placebo only in the
subgroup of rs2832407 C-allele homozygotes.
In a recent review, Guglilmo et al. (2015) concluded that topiramate shows a greater
beneficial effect in patients with a typology of craving, characterized by drinking obsessions and
automaticity of drinking. The group recommended topiramate (75-300 mg/d) as a first-line
treatment option for alcohol use disorders. A Cochrane review by Pani et al (2014) confirmed the
effects of topiramate on alcohol intake.
Topiramate may be used off label as a second-to-third-line medication in otherwise
treatment-refractory people.

9
ACCEPTED MANUSCRIPT

3.3.3. Gabapentin
Gabapentin is used and approved for the treatment of epilepsy and neuropathic pain. The drug
inhibits presynaptic voltage-gated Na+ and Ca2+ channels (Rogawski and Loscher, 2004) and

T
prevents the release of various neurotransmitters, including glutamate (Bonnet et al., 1999;

IP
Bozikas et al., 2002; Myrick et al., 1998; Rustembegovic et al., 2004; Watson et al., 1997). It may
reduce CNS hyperexcitability and anxiety during alcohol withdrawal (Watson et al., 1997).

R
The database on gabapentin in alcohol use disorders is limited. In a recent Cochrane

SC
review (Pani et al., 2014), it was found to have statistically significant positive effects on heavy
drinking (n=183 participants).

3.3.4. Baclofen
NU
Baclofen was promoted by the deceased cardiologist Olivier Ameisen, who reported and
MA
published his experiences of using it in to treat his own alcohol dependence (Ameisen, 2005;
2008). It is an agonist at the B subunit of GABA receptor (GABA-B) and was originally
D

approved for use in spasticity associated with neurologic disorders (for a review see Soyka and
Lieb, 2015). GABA neurotransmission is also known to be an important factor in the control of
TE

anxiety. Following these lines of evidence, activation of GABA-B receptors might reduce anxiety
(Addolorato et al., 2009). Baclofen inhibits dopaminergic neurotransmission via GABA-B
P

activation (Addolorato and Leggio, 2010).


CE

A series of RCTs conducted in Italy and an RCT conducted in the US generated


conflicting results for 30 mg/day baclofen (Addolorato et al., 2000; Addolorato et al., 2002;
AC

Addolorato et al., 2011; Addolorato et al., 2007; Garbutt et al., 2010; Morley et al., 2014).
Since baclofen has a low ability to cross the blood brain barrier, dosing may be a crucial
issue, because the drug must be titrated to reach higher doses. Previous studies used baclofen in
doses of 30-80 mg. An important but small RCT of high-dose baclofen up to 270 mg, funded by
the Deutsche Forschungsgemeinschaft, was published recently (N=56, Muller et al., 2015); this
study showed a higher abstinence rate and abstinence duration during the high-dose phase. Two
important French studies are currently under way. Baclofen has a temporary approval in France
for the treatment of alcohol-dependent patients for dosages up to 300 mg/d (see Muller et al.,
2015). In contrast to the positive findings, a recent 12-week RCT (N=64, Ponizovsky et al.,
2015) showed negative results. The use of baclofen in this area is not undisputed (Akosile and
Klan, 2015). Safety issues and interactions with alcohol, for example, are of relevance. Sedation
may be an important side effect: Clinical data indicate that the level of sedation in alcohol-

10
ACCEPTED MANUSCRIPT

dependent patients appears directly depended on the immediate doses of alcohol and baclofen
(Rolland et al., 2015). Baclofen has been viewed as a partial substitution in alcoholism (Rolland
et al., 2013). However, withdrawal symptoms or alcohol-like effects have not been reported in
the relevant studies (Addolorato et al., 2002; Garbutt et al., 2010; Muller et al., 2015). Anti-

T
craving effects have been described (Imbert et al., 2015). On the basis of the relatively high rate

IP
of acceptance of baclofen in patients with alcoholism, influenced by the Ameisen monography
and expressed in “baclofen forums” on the web, the drug may be viewed as an alternative strategy

R
in otherwise treatment-refractory patients with alcoholism with the main goal of abstinence.

SC
3.3.5. Varenicline

NU
Most alcohol-dependent people also smoke (Ait-Daoud et al., 2006; Nocente et al., 2013).
Varenicline is an approved medication for smoking cessation and was reported to lower alcohol-
consumption in smokers (Mitchell et al., 2012); for a review, see Erwin et al. (2014).
MA
Varenicline is a partial agonist at the α4β2 and full agonist at the α7 nicotinic
acetylcholine receptor in the ventral tegmental area, indirectly modulating dopaminergic
D

pathways (Crunelle et al., 2010; Crunelle et al., 2011; Muller et al., 2014; Nocente et al., 2013). It
probably modulates rewarding effects associated with alcohol intake (Ait-Daoud et al., 2006;
TE

Crunelle et al., 2010; Crunelle et al., 2011; Litten et al., 2013; Mitchell et al., 2012; Nocente et
al., 2013). Nausea is a common adverse effect (Erwin and Slaton, 2014). In a recent systematic
P

review, Erwin and Slaton (2014) identified 7 double-blind, placebo-controlled RCTs and 1 open-
CE

label study assessing the efficacy of varenicline on alcohol intake. In brief, there is some modest
evidence for improved in alcohol consumption, not only in smokers (Plebani et al., 2013).
AC

The data suggest that varenicline does not increase the percentage of days on which
participants abstain from alcohol but rather limits alcohol consumption after relapsing to alcohol.
The most commonly reported adverse effects are nausea, abnormal dreams, and constipation, all
of which are generally mild (Erwin and Slaton, 2014).
To date, the drug may be recommended for smokers who also want to quit alcohol.

3.3.6. Other medications


In Austria and Italy, sodium oxybate (gamma-hydroxy butyric acid, GHB) is approved for the
treatment of alcohol withdrawal syndrome and alcohol dependence (Keating, 2014). The drug
(“liquid ecstasy”) is a GABA-B agonist and has a significant risk for abuse, diversion, and use as
a “rape drug.” Few studies have been performed in alcohol withdrawal syndrome, but craving for
and abuse of sodium oxybate have been reported in alcohol use disorders (Skala et al., 2014).

11
ACCEPTED MANUSCRIPT

Because of its apparent risks (addictions), the drug cannot be recommended for widespread use in
alcoholism. The WHO (2012) warns against the use of GHB.
A few larger studies have been conducted on the serotonin 5-HT3 receptor antagonist
ondansetron. Johnson et al. (2000) performed a double-blind placebo-controlled RCT in 321

T
patients with alcoholism (271 randomized) and found that drinking was reduced only patients

IP
with early-onset alcoholism. Johnson et al. (2011) reported also that the serotonin transporter
gene may be of relevance for the effects of ondansetron. Individuals with the LL genotype who

R
received ondansetron had a lower mean number of drinks and a higher percentage of abstinent

SC
days than those who received placebo.
The neuroleptic aripiprazole, a partial D2, D3, and 5-HT1a agonist and 5-HT2a

NU
antagonist, may be of future interest for alcohol use disorders (Anton et al., 2008a; Edwards et
al., 2011). Aripiprazole may reduce alcohol craving and increase abstinence rates, as shown in a
clinical laboratory paradigm (Voronin et al., 2008). Anton et al. (2008a) failed to demonstrate
MA
superiority of aripiprazole over placebo in the primary endpoints (alcohol consumption, only on
subjective treatment effects).
D

Because stress tolerance, cortisol dysregulation, and the HPA stress axis and related
systems play a very significant role in the development of alcoholism (Emsley et al., 2013;
TE

Heilig and Koob, 2007), these systems may be targets for novel drugs in the treatment of
alcoholism. Corticotrophin releasing factor (CRF) antagonists or other drugs affecting
P

neuropeptide Y (NPY) or ghrelin are of potential interest. Clinical trials are ongoing with CRF 1
CE

antagonists in anxious alcoholics (Zorrilla et al., 2013). Again, the clinical perspective is vague.
AC

4. Opioid dependence
4.1 Approved medications
4.1.1 Opioid agonists
Opioid maintenance treatment is a well-established first-line approach for opioid dependence.
Methadone, buprenorphine, and the combination of buprenorphine and naloxone (in a 4:1 ratio)
are frequently used (Kleber et al., 2007; Lingford-Hughes et al., 2012; Mammen and Bell, 2009;
Mattick et al., 2009a; 2014; New South Wales Department of Health, 2006; Soyka, 2015; Soyka
et al., 2011a; Soyka et al., 2011b). These treatments have proven efficacy in reducing opioid
consumption and psychosocial and medical morbidity and in increasing treatment retention rates
and social functioning in people with opioid dependence (Mattick et al., 2009b; 2014).

4.1.2 Opioid antagonists

12
ACCEPTED MANUSCRIPT

Oral and (monthly) depot naltrexone are also available for the treatment of opioid-dependent
patients who are clearly abstinence oriented (Adi et al., 2007; Syed and Keating, 2013; Taylor et
al., 2011). However, both formulations are rarely used in opioid-dependent patients. Compliance
to treatment is rather low, at least for the oral formulation. Oral naltrexone appears to have some

T
limited benefit in helping formerly opioid-dependent individuals to remain abstinent, as stated in

IP
a comprehensive review by Adi et al. (2007). Depot naltrexone has been tested in Russia, where
methadone treatment is not available (Krupitsky et al., 2011). During or after naltrexone

R
treatment, patients no longer have an increased opioid tolerance, which may result in relapse to

SC
heroin, opioid overdose, and fatal intoxications. Other concerns include liver damage (Taylor et
al., 2011). Novel opioid antagonists are currently being developed and tested (Giuliano et al.,

NU
2015).

4.2 Alternative and novel treatments


MA
Most importantly, the dose of the agonist treatment must be adequate, because doses of
methadone or buprenorphine that are too low often provoke relapse to opioid use. Some alternate
D

pharmacological treatments are available for treatment-refractory patients who do not respond to
methadone or buprenorphine treatments.
TE

To improve compliance, depot or implant formulations of buprenorphine are currently


being developed. They have been tested in RCTs (Ling et al., 2010; Rosenthal et al., 2013) but
P

are not yet approved.


CE

A possible alternative treatment is an injectable opiate, either methadone or heroin


(diacetylmorphine). This approach has been studied predominantly in the UK in patients
AC

“persistently failing in orthodox treatment,” with encouraging results (Strang et al., 2010). Client
satisfaction seems to be quite good with this kind of approach (Groshkova et al., 2013). Both
injectable methadone and injectable heroin were found to be more cost effective than
optimized oral methadone in chronic refractory heroin users (Byford et al., 2013). In several
European countries, heroin maintenance is legal for patients not responding to conventional
treatment. A number of RCTs and clinical studies have been conducted, 6 of which were
included in a recent meta-analysis (Strang et al., 2015); see also Dursteler et al. (2015). Injectable
doses typically range between 150-250 mg per injection. The authors concluded that the overall
outcome improved in patients who did not respond to oral methadone treatment. An earlier
Cochrane analysis on this issue (Ferri et al., 2011) included 8 studies involving a total of 2007
patients. The analysis concluded that heroin has an added value alongside flexible doses of
methadone for long-term, treatment-refractory opioid users but also a higher rate of serious

13
ACCEPTED MANUSCRIPT

adverse events. Because injectable heroin treatment is less safe than conventional agonist
maintenance treatment and requires more clinical attention, its use is clearly limited to a small
subgroup of patients and can be considered a second-line option for treatment-refractory patients
(Garcia-Portilla et al., 2014). Legal regulations for heroin use vary significantly between

T
countries.

IP
Oral slow-release morphine is another treatment option. The drug has been studied as an
alternative to methadone for opioid detoxification (Madlung-Kratzer et al., 2009) and in

R
particular maintenance (Beck et al., 2014; Hammig et al., 2014). The Cochrane review was based

SC
on 3 RCTs only (Ferri et al., 2013). The authors stated that none of the studies included people
with a documented poor response to other maintenance treatment and that medical adverse events

NU
were consistently higher in people in slow-release morphine treatment. The drug is available in
some European countries and may be an alternative treatment for patients not responding to or
tolerating methadone because of side effects such as QTc prolongation.
MA

5. Cocaine Addiction
D

5.1 New pharmacological developments


Cocaine abuse and dependence have a wide range of adverse somatic and psychiatric health
TE

consequences (Kaye and Darke, 2004; Marzuk et al., 1998; Marzuk et al., 1992). However, there
is still no specific approved pharmacological treatment for cocaine addiction, although recent
P

advances in the neurobiology and pathophysiology of cocaine addiction have suggested several
CE

promising pharmacodynamic targets (Hulka et al., 2014; Kalivas and Volkow, 2011; Preti, 2007).
In addition to modafinil, methylphenidate, and disulfiram, which so far have the best
AC

evidence for the pharmacological treatment of cocaine addiction and thus are discussed in
more detail below, other medications have been tested for cocaine dependence in clinical
studies. Phase II and III studies have provided first evidence also for levodopa-carbidopa
and dextroamphetamine (dopaminergic agents); doxazosin and nepicastat (SYN 117;
noradrenergic agents); and topiramate, baclofen, N-acetylcysteine, and vigabatrin
(GABAergic agents) (Shorter et al., 2015). Finally, a cocaine vaccine (TA-CD) has been
investigated as a novel immunologic medication in a Phase III study for the treatment of
cocaine dependence (Kosten et al., 2014). However, this study did not find a significant
reduction in cocaine use among those receiving vaccination, compared to the placebo group.

5.1.1 Disulfiram

14
ACCEPTED MANUSCRIPT

Despite the fact that conclusive evidence supporting one specific compound is still lacking, at the
present time disulfiram seems to be the most promising agent for the treatment of cocaine
addiction (Suh et al., 2006). This conclusion is supported by about a dozen methodologically
sound RCTs of disulfiram use in cocaine-dependent patients (Baker et al., 2007; Carroll et al.,

T
2004; Carroll et al., 2000; Carroll et al., 1998; Grassi et al., 2007; Kosten et al., 2013; McCance-

IP
Katz et al., 1998; Oliveto et al., 2011; Petrakis et al., 2000; Pettinati et al., 2008; Schottenfeld et
al., 2014; Shorter et al., 2013; Spellicy et al., 2013). Besides the inhibition of aldehyde

R
dehydrogenase (see 3.2.1), inhibition of brain DA-beta-hydroxylase (DBH) and subsequent

SC
monoaminergic modulation has been discussed recently as another possible direct
pharmacological effect in the central nervous system (Faiman et al., 2013; Mutschler et al., 2009;

NU
Weinshenker, 2010). These significant changes in dopaminergic and noradrenergic
neurotransmission have been suggested to be the pharmacological mechanism of action of
disulfiram in the treatment of addictive disorders such as cocaine dependence and pathological
MA
gambling (Mutschler et al., 2010; Mutschler et al., 2009). However, it is unclear whether the
inhibition of DBH reported in animal studies leads to clinically relevant changes in dopaminergic
D

transmission in the human brain. Furthermore, disulfiram is not specifically classified as a


stimulant medication, although dopaminergic and noradrenergic changes as a direct consequence
TE

of acute disulfiram administration have been shown in animal studies. In clinical studies,
disulfiram has shown initial promise in the treatment of cocaine addiction alone and in patients
P

with a dual diagnosis of substance abuse, e.g., alcohol and cocaine.


CE

5.1.2 Methylphenidate
AC

Methylphenidate (MPH), when used as a stimulant medication, has been shown to have potential
as a treatment for cocaine addiction (Dursteler, 2015). The initial idea behind the use of MPH and
other stimulant medications (such as modafinil) in the treatment of cocaine addiction was based
on the replacement-substitution therapy approach (Dursteler, 2015). This approach has been very
successful in reducing drug-related harm in treatments for opioid and nicotine dependence (Batra,
2011; Nordt and Stohler, 2009).
Both cocaine and MPH target the DA system by inhibiting the presynaptic DA
transporters and thus have very similar pharmacodynamic properties (Calipari et al., 2014). Like
cocaine, MPH acts as a monoamine reuptake inhibitor, thereby increasing extracellular
catecholamine concentrations (Calipari et al., 2014). Inhibition of the presynaptic DA transporters
results in increased extracellular DA levels within the mesolimbic reward system (Ritz et al.,
1987). However, cocaine dependence not only alters the DA system; the reinforcing effects of

15
ACCEPTED MANUSCRIPT

cocaine are mediated also via serotonin, norepinephrine, and other neurotransmitters (Hulka et
al., 2014; Rocha, 2003; Weinshenker and Schroeder, 2007).
MPH is approved and mainly indicated for the treatment of attention-
deficit/hyperactivity disorder (ADHD), but it is used also in the treatment of excessive daytime

T
sleepiness and narcolepsy (Epstein et al., 2014; Leonard et al., 2004). When used as indicated,

IP
MPH is well tolerated and has a good safety profile, as reported in many studies (Epstein et al.,
2014). However, MPH is known also to have reinforcing effects similar to those of cocaine

R
(Kollins et al., 2001; Parran and Jasinski, 1991). This means that MPH has a potential for abuse

SC
and dependence and thus has been classified as a schedule II controlled substance in the USA.
The illicit use of MPH has also expanded because of (mis)use as a cognitive enhancer to improve

NU
intellectual performance (Maier et al., 2015).
To date, only a small number of studies have considered MPH as a treatment for
cocaine-dependent patients. Notably, positive studies have primarily consisted of case reports and
MA
open-label trials. A recently published review by Dürsteler et al. (2015) found that 4 out of 5
published case reports and all 3 open-label studies (N=83) reported positive effects for MPH. By
D

contrast, investigation of the effects of MPH on cocaine dependence in the 5 double-blind


placebo-controlled RCTs published to date produced a quite different picture: 4 out of 5 found no
TE

benefit for MPH compared to placebo (N=363; Dursteler, 2015). Only one study, by Levin et al.
(2007), reported a reduced likelihood of cocaine use over time in cocaine-dependent patients with
P

comorbid ADHD.
CE

In summary, current evidence for the use of MPH in the treatment of cocaine abuse and
dependence is still very weak, and such evidence as there is derives mainly from case reports.
AC

Case studies tend to focus on positive outcomes and show other methodological limitations,
meaning that general conclusions on possible effects cannot be derived from this type of study.
Studies with a high level of scientific evidence (RCTs) failed to find significant benefits for MPH
in reducing cocaine use compared to placebo. In addition, the reported risks of MPH beyond the
approved usage suggest potential for abuse, due to both the reinforcing effects of MPH and the
performance-enhancing effects on cognition. An increasing number of students reportedly abuse
MPH, and some patients show MPH dependence (Bjarnadottir et al., 2015; Gahr et al., 2014;
Peles et al., 2015). A recent study by Harel-Fisch et al. (2013) amongst youth aged 17-18 years
found a 0.9 % rate of non-prescribed use of MPH. Much higher rates of MPH abuse were
reported by Peles et al. (2015) in a recent cross-sectional study of patients on methadone
maintenance treatment. These investigators found a 14.7% (n=45/306) prevalence of MPH abuse
in urine tests in Tel Aviv. However, in the same study none of the 190 patients tested in Las

16
ACCEPTED MANUSCRIPT

Vegas showed positive MPH results (Peles et al., 2015). Potential contributory factors to high
rates of MPH use could be easy accessibility or the high MPH prescription rates seen in some
countries. These striking differences in reported misuse of MPH call for further research to better
understand the factors contributing to national variation and higher rates of MPH abuse in certain

T
countries.

IP
Prospective studies have reported an elevated comorbidity of substance dependence and
ADHD (van de Glind et al., 2014; Wilens et al., 1994). Because of the overlapping

R
pathophysiological aspects of both disorders (e.g. dopaminergic deficits, executive dysfunction,

SC
common genetic risk factors), some patients may self-treat their ADHD symptoms with cocaine
(“self-medication theory”) (Khantzian, 1983). This comorbidity suggests that patients with

NU
ADHD are more vulnerable to the development of substance dependence. Treatment of ADHD
may very well reduce the risk for substance abuse or dependence (Paslakis et al., 2010).
However, MPH study results are unambiguous in this regard: A recent meta-analysis of 15
MA
studies found no protective effects of MPH on the development of later SUDs (Humphreys et al.,
2013).
D

Thus far, it is unclear whether particular subgroups of cocaine-dependent patients show a


better response to MPH, although leading candidates may be patients with comorbid ADHD. We
TE

therefore suggest that pharmacological substitution therapy with MPH should be considered for
treatment-resistant cocaine dependence and comorbid ADHD (Paslakis et al., 2010). Treating
P

physicians should regularly interview and monitor patients regarding their condition (e.g. craving,
CE

consumption), with a particular emphasis on increasing the dosage of MPH. The evidence
supporting MPH treatment of cocaine-dependent patients without comorbid ADHD is in our
AC

opinion too weak to allow recommendation of the therapy and would probably even be dangerous
in some cases. Thus, we recommend caution regarding MPH use when treating patients with a
history of cocaine dependence.

5.1.3 Modafinil
Modafinil is another promising stimulant medication for the treatment of cocaine abuse and
dependence. Modafinil is an alpha-adrenergic/glutamate agonist that inhibits DA re-uptake. It is
an approved treatment for narcolepsy, shift work sleep disorder, and obstructive sleep apnea
syndrome. In 2 placebo-controlled RCTs in cocaine-dependent patients, modafinil (200 mg – 400
mg/day) was shown to reduce craving and increase cocaine abstinence compared to placebo (total
N=272) (Anderson et al., 2009; Dackis et al., 2005). In an open-label study (N=65) recently
published by Nuijten et al. (2015), adherence to modafinil was very low, with only a 10% rate of

17
ACCEPTED MANUSCRIPT

treatment completion. Furthermore, no significant benefit was found for modafinil as an add-on
treatment to cognitive behavioral therapy (Dackis et al., 2012). Taken together, the few studies
that have investigated modafinil as a treatment for cocaine dependence have produced
inconclusive outcomes. Further research is warranted to identify possible modafinil

T
responders/non-responders in the treatment of cocaine dependence. Finally, it is important to

IP
keep in mind that modafinil is listed as a schedule IV drug in the United States and that the
medication is a potent reinforcer at higher dosages. First case reports of modafinil-associated

R
psychosis, withdrawal symptoms, and abuse/dependence have also been reported (Kate et al.,

SC
2012; Krishnan and Chary, 2015; Mariani and Hart, 2005).

NU
6. Pharmacogenetics
Pharmacogenetic approaches may help to optimize treatment response in otherwise treatment-
refractory patients and could represent an important strategy to further improve treatment
MA
effectiveness by allowing responders/non-responders to be identified.
A118G, a common and clinically relevant single nucleotide polymorphism of the
D

OPRM1 gene at the mu-opioid receptor, results in an amino acid exchange at position 40 from
asparagine to aspartate (Bond et al., 1998). The OPRM1 118G genotype appears to moderate the
TE

subjective and neuronal response of opioid antagonists to alcohol and alcohol cue reactivity
(Ashenhurst et al., 2012; Schacht et al., 2013; Setiawan et al., 2012) and treatment response with
P

opioid antagonists. Results for naltrexone are mixed and were obtained mainly from post hoc
CE

analyses (Anton et al., 2008b; Anton et al., 2012; Gelernter et al., 2007; Kim et al., 2009;
Kranzler et al., 2013; Oroszi et al., 2009). A meta-analysis stressed the role of the A118G
AC

polymorphism of the OPRM1 gene in moderating the effect of naltrexone in patients with alcohol
dependence and treatment response (Chamorro et al., 2012), but there are recent negative findings
(Oslin et al., 2015).
Recently, promising pharmacogenetic studies were published showing an influence of
genetic polymorphisms (e.g., DBH, ADRA1A genes) on the outcome of disulfiram treatment in
cocaine-dependent patients (Kosten et al., 2013; Shorter et al., 2013; Spellicy et al., 2013).

7. Neuromodulation: Repeated Transcranial Magnetic Stimulation and Deep Brain


Stimulation in Substance Use Disorders
SUDs are the result of a complex interaction of genes, environment, and substance effects. As
described above, a variety of treatment options are currently available for patients with SUDs.
However, only a small percentage of patients treated for SUDs remain abstinent in the long term,

18
ACCEPTED MANUSCRIPT

and patients often develop a chronic course with multiple relapses. In one study, only 6% to 30%
of alcohol-dependent patients remained abstinent 2 to 3 years after treatment (Project MATCH
Research Group, 1998). Without further treatment after inpatient detoxification, most patients
with SUDs relapse after discharge, and this relapse is relatively independent of the substance or

T
drug used (e.g. opioids, cocaine, nicotine, or alcohol) (Berglund et al., 2003; Dutra et al., 2008).

IP
One of the underlying causes of relapse might relate to the broad clinical heterogeneity of SUDs,
in terms of symptom dimensions, severity, and psychiatric comorbidity, and the wide variability

R
in treatment response. Almost all effective therapeutic interventions interact with the brain

SC
structures that are known to be involved in reward and motivation pathways. Improving treatment
results calls for effective treatment approaches, especially for SUD patients with multiple relapses

NU
and a chronic course of addiction. New and innovative electrophysiological treatment options,
including deep brain stimulation (DBS) and repeated transcranial magnetic stimulation (rTMS),
show potential in the direct modulation of dysfunctional brain networks in patients with
MA
(treatment-refractory) SUDs and may represent a novel tool to supplement pharmacological and
psychosocial/psychotherapeutic approaches.
D
TE

7.1 Deep brain stimulation (DBS)


DBS is a neurosurgical technique with proven effectiveness as an additional therapeutic
intervention in the treatment of severe neurological movement disorders (e.g. Parkinson disease,
P

dystonia, and essential tremor) (Kern and Kumar, 2007). It involves the delivery of electrical
CE

pulses via electrodes implanted in defined regions of the brain, which results in direct interactions
with neurons and cortico-striatal circuits. Despite the fact that the exact mechanisms of action of
AC

DBS still are largely unknown, the growing interest worldwide in DBS for SUDs has resulted in
increasing numbers of publications on the topic. Although still less widely investigated and
implemented than in the field of movement disorders, the application of DBS in psychiatric
illnesses such as depression, schizophrenia, anorexia, obsessive-compulsive disorder, therapy-
resistant Tourette syndrome, and drug addiction has also shown promising results (Bartsch and
Kuhn, 2014; Coenen et al., 2015; de Haan et al., 2015; Greenberg et al., 2008; Kuhn et al.,
2011a; Luigjes et al., 2012; Stephen et al., 2012; Sun et al., 2015; Voges et al., 2013). The
majority of studies of DBS in psychiatric illness have focused on the treatment of therapy-
resistant obsessive-compulsive disorder (OCD) (Greenberg et al., 2008). In this section, we
provide a summary of the available clinical evidence on DBS treatment of patients with SUDs.
The application of DBS in SUDs generally remains at the stage of an experimental
therapy, without official approval. DBS was originally developed in animal research; the only

19
ACCEPTED MANUSCRIPT

evidence in humans derived from case reports (Kravitz et al., 2015). The earliest studies of DBS
were in the 1950s, involving experiments on self-stimulation in rats in which animals with
implanted electrodes robustly sought stimulation of the septal region, resulting in apparent
feelings of pleasure (Olds and Milner, 1954). Subsequent preclinical studies investigated the

T
effects of DBS in animal models of addiction to ethanol (Knapp et al., 2009), morphine (Liu et

IP
al., 2008), and cocaine (Rouaud et al., 2010), affecting target brain areas that included the
subthalamic nucleus, lateral habenula, medial prefrontal cortex, lateral hypothalamus, and

R
nucleus accumbens. In a review of DBS in animal studies, Luigjes et al. (2012) concluded that

SC
stimulation of the nucleus accumbens showed the most robust effects in different models of
addiction.

NU
To date, only a handful of human studies have been published that describe the use of
DBS in patients with SUDs. In most of these publications, patients were treated with DBS
primarily in relation to other conditions such as Parkinson’s disease, anxiety disorders, or OCD.
MA
The positive effects on addiction were generally observed secondarily during follow-up. The
most commonly targeted brain areas in patients with SUD were the nucleus accumbens and the
D

subthalamic nucleus. Positive therapeutic effects of DBS on these brain regions have been
described for addictions to alcohol (Kuhn et al., 2011b; Kuhn et al., 2007; Muller et al., 2009;
TE

Voges et al., 2013), nicotine (Kuhn et al., 2009; Mantione et al., 2010; Zhou et al., 2011), and
heroin (Stephen et al., 2012; Valencia-Alfonso et al., 2012; Zhou et al., 2011) and for behavioral
P

addictions (Ardouin et al., 2006; Bandini et al., 2007). DBS studies in patients with SUDs are
CE

summarized briefly in Table 1.


Despite these initial promising results, additional studies are required before definitive
AC

conclusions can be reached on the efficacy and safety of DBS in SUDs. Almost all currently
available studies on DBS in SUDs have clear limitations, because they were non-randomized,
open-label studies or small case-control series. In some patients, SUDs actually worsened after
DBS. Besides the obvious questions regarding methodological and ethical aspects and regarding
the presently unknown long-term effects of DBS, improving understanding of treatment-resistant
SUDs is also a priority. Available reports also differ in terms of stimulation parameters and
targeted brain areas, emphasizing the need for further research on the application of DBS in
patients with SUDs before these outstanding questions can be resolved.
Another important challenge involves the recruitment of patients with SUDs for DBS
RCTs. One recent report described this as being much more difficult than recruiting patients with
OCD (Luigjes et al., 2015). Two registered DBS RCTs are currently recruiting SUD patients
(Cologne, Germany, and Amsterdam, Netherlands), but both research groups are experiencing

20
ACCEPTED MANUSCRIPT

problems in obtaining sufficient numbers of patients, and it is not clear whether these studies will
eventually recruit the originally envisaged number of study participants (Luigjes et al., 2015).
These problems mean that current evidence supporting DBS is limited to that derived from
preclinical studies, human case reports, or case series. However, despite these reservations,

T
patients with treatment-refractory SUDs could be eligible for DBS as a possible therapeutic

IP
option in severe cases (preferably within the setting of an RCT), thus effectively extending
current therapeutic options in patients with SUDs.

R
SC
7.2 Repeated Transcranial Magnetic Stimulation (rTMS)
rTMS is another electrophysiological therapeutic approach to treating patients with SUDs

NU
(Bellamoli et al., 2014; Sousa, 2013). In contrast to DBS, rTMS is a non-invasive method with
many advantages over DBS in the treatment of patients with SUDs that therefore has much
greater potential to become an established therapy for SUDs (Bellamoli et al., 2014; Sousa,
MA
2013). rTMS is based on magnetic field pulses delivered via a stimulation coil close to the head;
the magnetic field induces an electrical field in the brain, thereby activating cortical neurons
D

(Fitzgerald et al., 2009). rTMS can induce sustained changes in cortical excitability and brain
activities and may affect the HPA axis (Baeken et al., 2009). Furthermore, rTMS has been shown
TE

to induce DA release in subcortical and cortical brain areas (Strafella et al., 2001). The main brain
target for rTMS in the treatment of SUDs is the prefrontal cortical network, in particular the
P

dorsolateral prefrontal cortex (DLPFC) and the orbitofrontal cortex (Gorelick et al., 2014;
CE

Mishra et al., 2015). These brain regions have important functions in the inhibitory control of
addictive substances, a mechanism often impaired in patients with SUDs (Herremans et al.,
AC

2015; Kravitz et al., 2015; Prikryl et al., 2014; Protasio et al., 2015). Increased activity of the
prefrontal cortex could be one major mechanism of action of rTMS in patients with SUDs (Feil
and Zangen, 2010). Other possible mechanisms of action include the reduction of craving, the
modulation of the dopaminergic and HPA systems, and the modulation of decision-making
processes, leading to a reduction in risk-taking behavior (Knoch et al., 2006).
Many studies have been published on rTMS, and the technique is considered safe
(Levkovitz et al., 2007; Rossi et al., 2009). Depending on the location and parameters of
stimulation, a range of changes in behavior can be induced. High-frequency rTMS (5-20 Hz) is
thought to increase synaptic transmission, whereas low-frequency rTMS (0.2-1 Hz) is thought to
decrease synaptic transmission (Fitzgerald et al., 2006). Repetition of TMS is required to induce
the long-term synaptic changes that are important for prolonged therapeutic effects, and effects
may last for weeks (Fitzgerald et al., 2006). rTMS can be applied safely and painlessly to

21
ACCEPTED MANUSCRIPT

patients, and the technique can be used in addition to other treatments (as an augmentation
therapy). rTMS is rarely the only (standard) treatment.
Despite modest therapeutic effects in the majority of studies, rTMS reportedly achieves
therapeutic-like effects in major depression. Consequently, the FDA recently approved rTMS for

T
depressed patients (Daskalakis et al., 2008). In principle, any psychiatric disorder that includes

IP
cortical dysfunction may benefit from rTMS. Therefore, rTMS has been investigated in
conditions such as mania, OCD, PTSD, and schizophrenia (Burt et al., 2002; Dunlop et al.,

R
2015; Grammer et al., 2015; Hasan et al., 2015).

SC
To date, studies on the application of rTMS for SUDs have most frequently investigated
alcohol, nicotine, and cocaine SUDs (Table 2 gives a brief summary of the studies on alcohol

NU
and cocaine). In addition, preliminary reports of promising rTMS interventions in other addictive
diseases, including opiates, 3,4-methylenedioxymethamphetamine (MDMA), and gambling (Barr
et al., 2011), also have been published recently. Taken together, studies on rTMS as a treatment
MA
for SUDs suggest efficacy, particularly in the reduction of craving in patients dependent on
alcohol, nicotine, and cocaine. Furthermore, there seems to be a potential for high frequency
D

rTMS to reduce not only craving, but also the consumption of alcohol, nicotine, and cocaine.
However, the limitations of the available studies should be recognized, the main issues being the
TE

absence of reporting of long-term effects in most studies and the short-lived nature of effects on
craving and consumption. Moreover, some studies reported inconsistent outcomes for craving and
P

consumption measures, illustrating the difficulties in the operationalization of the concept


CE

“craving” and the verification of “consumption.” Objective measures of consumption, e.g. hair
analysis for drugs (Madry et al., 2014), could provide better evidence for the effectiveness of
AC

rTMS in SUDs. Furthermore, studies differed in technical and stimulation standards, making it
difficult to comment on the optimal hemisphere and rTMS parameters for the treatment of SUDs.
The studies reviewed in Table 2 support the efficacy of high frequency rTMS stimulation of the
right and/or left DLPFC. Further studies should investigate larger sample sizes, identify ideal
stimulation standards and, finally, investigate rTMS in further SUDs.
In conclusion, rTMS is a safe, non-invasive therapeutic method with demonstrably
positive effects on craving and consumption in patients with SUDs. In addition, rTMS could be
easily applied together with other SUD treatments as an augmentation therapy, without a risk of
side effects.

Conclusion

22
ACCEPTED MANUSCRIPT

Treatment-refractory substance use disorders require multidimensional therapeutic


approaches. In this review, we focus on novel pharmacological and brain stimulation
techniques that have the potential to enrich treatment outcomes, at least for some patients.
However, pharmacological and stimulation techniques always have to be combined with

T
psychosocial/psychotherapeutic treatments. Further studies are needed to find new

IP
therapeutic strategies/targets and to further elucidate the discussed treatment options.

R
Acknowledgements

SC
The authors thank Jacquie Klesing, Board-certified Editor in the Life Sciences (ELS), and Dr. J.
P. Bayley (Medactie), for editing assistance with the manuscript.

Statement of interest
NU
No funding was used to prepare this narrative. For the past 5 years, M.S. has worked as a
MA
consultant or has received research grants from Sanofi Aventis, Novartis, Mepha, Reckitt
Benckiser, and Lundbeck. J.M. has received travel expenses and consultant fees from Lundbeck
D

and Takeda.
P TE
CE
AC

23
ACCEPTED MANUSCRIPT

Table 1
Studies of Deep Brain Stimulation in Substance Use Disorders

Target brain Stimulation


Reference N Substance DBS effects
area Frequency
Mantione et al.
1 nicotine Bilateral NAc 180 Hz 1 resolved

T
(2010)
Kuhn et al. 5 bilateral, 5 3 resolved, 7

IP
10 nicotine 130-145 Hz
(2007) unilateral NAc unchanged
Müller et al. 2 resolved, 1

R
3 alcohol Bilateral NAc 130 Hz
(2010) improved
Kuhn et al.

SC
1 alcohol Bilateral NAc 130 Hz 1 improved
(2009)
Zhou et al.
1 heroin Bilateral NAc 145 Hz 1 resolved
(2011)

NAc: Nucleus accumbens


NU
MA
D
TE
P
CE
AC

24
ACCEPTED MANUSCRIPT

Table 2
Studies of repeated transcranial magnetic stimulation (rTMS) in Substance Use Disorders

Target
Stimulation Sham
Reference N Substance brain rTMS effects

T
frequency condition
area

IP
Reduction in
Ceccanti et
18 alcohol MLPFC 20 Hz Yes craving and
al. (2015)
alcohol intake

R
No effect on

SC
Herremans et Right immediate or
36 alcohol 20 Hz Yes
al. (2012) DLPFC long-term
craving
Höppner et 10, 9 Left No reduction

NU
alcohol 20 Hz Yes
al. (2011) (females) DLPFC in craving
Reduction in
immediate
MA
Mishra et al. Right craving, but
30, 15 alcohol 10 Hz Yes
(2010) DLPFC no effect on
craving after
4 weeks
D

Yes (right Reduction in


Mishra et al.
20 alcohol DLPFC 10 Hz vs. left craving in
TE

(2015)
DLPFC) both groups
Reduction in
craving and
P

1 (48-year- consumption
de Ridder et
CE

old alcohol dACC 1 Hz No (but relapse


al. (2011)
woman) after 3 months
with increased
craving)
AC

Reduction in
craving (right
DLPFC),
reduction in
anxiety,
Right and increase in
Camprodon
6 cocaine left 10 Hz No mood after
et al. (2007)
DLPFC right-sided
and in
unhappiness
after left-
sided
stimulation
0.03–0.08 Increased safe
Yes (right
Gorini et al. mA/cm2 behavior after
18,18 cocaine DLPFC vs. left
(2014) when used right DLPFC
DLPFC)
with a current stimulation,

25
ACCEPTED MANUSCRIPT

of 1.5 mA risk-taking
behavior
increased
after left
DLPFC
stimulation

T
Politi et al. Left Reduction in
36 cocaine 15 Hz No
(2008) DLPFC craving

IP
DLPFC: Dorsolateral prefrontal cortex; dACC: Dorsal anterior cingulated cortex; MLPFC:

R
Medial prefrontal cortex

SC
NU
MA
D
P TE
CE
AC

26
ACCEPTED MANUSCRIPT

References
Addolorato G, Caputo F, Capristo E, Colombo G, Gessa GL, Gasbarrini G. Ability of baclofen
in reducing alcohol craving and intake: II--Preliminary clinical evidence. Alcohol Clin
Exp Res 2000;24:67-71.
Addolorato G, Caputo F, Capristo E, Domenicali M, Bernardi M, Janiri L, et al. Baclofen
efficacy in reducing alcohol craving and intake: a preliminary double-blind
randomized controlled study. Alcohol Alcohol 2002;37:504-8.

T
Addolorato G, Leggio L. Safety and efficacy of baclofen in the treatment of alcohol-

IP
dependent patients. Curr Pharm Des 2010;16:2113-7.
Addolorato G, Leggio L, Cardone S, Ferrulli A, Gasbarrini G. Role of the GABA(B) receptor
system in alcoholism and stress: focus on clinical studies and treatment perspectives.

R
Alcohol 2009;43:559-63.
Addolorato G, Leggio L, Ferrulli A, Cardone S, Bedogni G, Caputo F, et al. Dose-response

SC
effect of baclofen in reducing daily alcohol intake in alcohol dependence: secondary
analysis of a randomized, double-blind, placebo-controlled trial. Alcohol Alcohol
2011;46:312-7.
Addolorato G, Leggio L, Ferrulli A, Cardone S, Vonghia L, Mirijello A, et al. Effectiveness and

NU
safety of baclofen for maintenance of alcohol abstinence in alcohol-dependent
patients with liver cirrhosis: randomised, double-blind controlled study. Lancet
2007;370:1915-22.
Adi Y, Juarez-Garcia A, Wang D, Jowett S, Frew E, Day E, et al. Oral naltrexone as a
MA
treatment for relapse prevention in formerly opioid-dependent drug users: a
systematic review and economic evaluation. Health Technol Assess 2007;11:iii-iv, 1-
85.
Ait-Daoud N, Lynch WJ, Penberthy JK, Breland AB, Marzani-Nissen GR, Johnson BA. Treating
D

smoking dependence in depressed alcoholics. Alcohol Res Health 2006;29:213-20.


Akosile W, Klan M. Creating a new problem: The use of baclofen in the management of
TE

alcohol use disorder. Drug Alcohol Rev 2015.


Ameisen O. Complete and prolonged suppression of symptoms and consequences of alcohol-
dependence using high-dose baclofen: a self-case report of a physician. Alcohol
Alcohol 2005;40:147-50.
P

Ameisen O. The End of My Addiction. New York: Farrar, Straus and Giroux; 2008.
Anderson AL, Reid MS, Li SH, Holmes T, Shemanski L, Slee A, et al. Modafinil for the
CE

treatment of cocaine dependence. Drug Alcohol Depend 2009;104:133-9.


Angehagen M, Ronnback L, Hansson E, Ben-Menachem E. Topiramate reduces AMPA-induced
Ca(2+) transients and inhibits GluR1 subunit phosphorylation in astrocytes from
AC

primary cultures. J Neurochem 2005;94:1124-30.


Anton RF, Kranzler H, Breder C, Marcus RN, Carson WH, Han J. A randomized, multicenter,
double-blind, placebo-controlled study of the efficacy and safety of aripiprazole for
the treatment of alcohol dependence. J Clin Psychopharmacol 2008a;28:5-12.
Anton RF, O'Malley SS, Ciraulo DA, Cisler RA, Couper D, Donovan DM, et al. Combined
pharmacotherapies and behavioral interventions for alcohol dependence: the
COMBINE study: a randomized controlled trial. Jama 2006;295:2003-17.
Anton RF, Oroszi G, O'Malley S, Couper D, Swift R, Pettinati H, et al. An evaluation of mu-
opioid receptor (OPRM1) as a predictor of naltrexone response in the treatment of
alcohol dependence: results from the Combined Pharmacotherapies and Behavioral
Interventions for Alcohol Dependence (COMBINE) study. Arch Gen Psychiatry
2008b;65:135-44.
Anton RF, Pettinati H, Zweben A, Kranzler HR, Johnson B, Bohn MJ, et al. A multi-site dose
ranging study of nalmefene in the treatment of alcohol dependence. J Clin
Psychopharmacol 2004;24:421-8.
Anton RF, Voronin KK, Randall PK, Myrick H, Tiffany A. Naltrexone modification of drinking
effects in a subacute treatment and bar-lab paradigm: influence of OPRM1 and
dopamine transporter (SLC6A3) genes. Alcohol Clin Exp Res 2012;36:2000-7.
APA. Diagnostic and statistical manual of mental disorders, 4th edition, Text Revision.
Washington, DC: American Psychiatric Publishing; 2000.

27
ACCEPTED MANUSCRIPT

APA. Diagnostic and statistical manual of mental disorders (5th ed.). Arlington, VA:
American Psychiatric Publishing; 2013.
Ardouin C, Voon V, Worbe Y, Abouazar N, Czernecki V, Hosseini H, et al. Pathological
gambling in Parkinson's disease improves on chronic subthalamic nucleus stimulation.
Movement disorders : official journal of the Movement Disorder Society
2006;21:1941-6.
Ashenhurst JR, Bujarski S, Ray LA. Delta and kappa opioid receptor polymorphisms influence

T
the effects of naltrexone on subjective responses to alcohol. Pharmacol Biochem
Behav 2012;103:253-9.

IP
Baeken C, De Raedt R, Leyman L, Schiettecatte J, Kaufman L, Poppe K, et al. The impact of
one HF-rTMS session on mood and salivary cortisol in treatment resistant unipolar
melancholic depressed patients. J Affect Disord 2009;113:100-8.

R
Baker JR, Jatlow P, McCance-Katz EF. Disulfiram effects on responses to intravenous cocaine
administration. Drug Alcohol Depend 2007;87:202-9.

SC
Bandini F, Primavera A, Pizzorno M, Cocito L. Using STN DBS and medication reduction as a
strategy to treat pathological gambling in Parkinson's disease. Parkinsonism & related
disorders 2007;13:369-71.

NU
Barr MS, Farzan F, Wing VC, George TP, Fitzgerald PB, Daskalakis ZJ. Repetitive transcranial
magnetic stimulation and drug addiction. International review of psychiatry
2011;23:454-66.
Bart G, Schluger JH, Borg L, Ho A, Bidlack JM, Kreek MJ. Nalmefene induced elevation in
MA
serum prolactin in normal human volunteers: partial kappa opioid agonist activity?
Neuropsychopharmacology 2005;30:2254-62.
Bartsch C, Kuhn J. [Deep brain stimulation for addiction, anorexia and compulsion.
Rationale, clinical results and ethical implications]. Nervenarzt 2014;85:162-8.
Batki SL, Pennington DL, Lasher B, Neylan TC, Metzler T, Waldrop A, et al. Topiramate
D

treatment of alcohol use disorder in veterans with posttraumatic stress disorder: a


randomized controlled pilot trial. Alcohol Clin Exp Res 2014;38:2169-77.
TE

Batra A. Treatment of tobacco dependence. Deutsches Arzteblatt international


2011;108:555-64.
Beck T, Haasen C, Verthein U, Walcher S, Schuler C, Backmund M, et al. Maintenance
P

treatment for opioid dependence with slow-release oral morphine: a randomized


cross-over, non-inferiority study versus methadone. Addiction 2014;109:617-26.
CE

Bellamoli E, Manganotti P, Schwartz RP, Rimondo C, Gomma M, Serpelloni G. rTMS in the


treatment of drug addiction: an update about human studies. Behavioural neurology
2014;2014:815215.
Bencherif B, Wand GS, McCaul ME, Kim YK, Ilgin N, Dannals RF, et al. Mu-opioid receptor
AC

binding measured by [11C]carfentanil positron emission tomography is related to


craving and mood in alcohol dependence. Biol Psychiatry 2004;55:255-62.
Berglund M, Thelander S, Jonsson E. Treating alcohol and drug abuse - an evidence based
review. Weinheim: Wiley-VCH Verlag GmbH & Co. KGaA; 2003.
Biton V, Edwards KR, Montouris GD, Sackellares JC, Harden CL, Kamin M. Topiramate
titration and tolerability. Ann Pharmacother 2001;35:173-9.
Bjarnadottir GD, Haraldsson HM, Rafnar BO, Sigurdsson E, Steingrimsson S, Johannsson M,
et al. Prevalent intravenous abuse of methylphenidate among treatment-seeking
patients with substance abuse disorders: a descriptive population-based study. J
Addict Med 2015;9:188-94.
Blodgett JC, Del Re AC, Maisel NC, Finney JW. A meta-analysis of topiramate's effects for
individuals with alcohol use disorders. Alcohol Clin Exp Res 2014;38:1481-8.
Bond C, LaForge KS, Tian M, Melia D, Zhang S, Borg L, et al. Single-nucleotide
polymorphism in the human mu opioid receptor gene alters beta-endorphin binding
and activity: possible implications for opiate addiction. Proc Natl Acad Sci U S A
1998;95:9608-13.
Bonnet U, Banger M, Leweke FM, Maschke M, Kowalski T, Gastpar M. Treatment of alcohol
withdrawal syndrome with gabapentin. Pharmacopsychiatry 1999;32:107-9.
Bozikas V, Petrikis P, Gamvrula K, Savvidou I, Karavatos A. Treatment of alcohol withdrawal
with gabapentin. Prog Neuropsychopharmacol Biol Psychiatry 2002;26:197-9.

28
ACCEPTED MANUSCRIPT

Braillon A. Nalmefene in alcohol misuse: junk evaluation by the European Medicines Agency.
Bmj 2014;348:g2017.
Burt T, Lisanby SH, Sackeim HA. Neuropsychiatric applications of transcranial magnetic
stimulation: a meta analysis. Int J Neuropsychopharmacol 2002;5:73-103.
Byford S, Barrett B, Metrebian N, Groshkova T, Cary M, Charles V, et al. Cost-effectiveness
of injectable opioid treatment v. oral methadone for chronic heroin addiction. Br J
Psychiatry 2013;203:341-9.

T
Calipari ES, Ferris MJ, Melchior JR, Bermejo K, Salahpour A, Roberts DC, et al.
Methylphenidate and cocaine self-administration produce distinct dopamine terminal

IP
alterations. Addict Biol 2014;19:145-55.
Carroll KM, Fenton LR, Ball SA, Nich C, Frankforter TL, Shi J, et al. Efficacy of disulfiram and
cognitive behavior therapy in cocaine-dependent outpatients: a randomized placebo-

R
controlled trial. Arch Gen Psychiatry 2004;61:264-72.
Carroll KM, Nich C, Ball SA, McCance E, Frankforter TL, Rounsaville BJ. One-year follow-up of

SC
disulfiram and psychotherapy for cocaine-alcohol users: sustained effects of
treatment. Addiction 2000;95:1335-49.
Carroll KM, Nich C, Ball SA, McCance E, Rounsavile BJ. Treatment of cocaine and alcohol

NU
dependence with psychotherapy and disulfiram. Addiction 1998;93:713-27.
Chamorro AJ, Marcos M, Miron-Canelo JA, Pastor I, Gonzalez-Sarmiento R, Laso FJ.
Association of micro-opioid receptor (OPRM1) gene polymorphism with response to
naltrexone in alcohol dependence: a systematic review and meta-analysis. Addict Biol
MA
2012;17:505-12.
Coenen VA, Amtage F, Volkmann J, Schlapfer TE. Deep Brain Stimulation in Neurological and
Psychiatric Disorders. Deutsches Arzteblatt international 2015;112:519-26.
Crunelle CL, Miller ML, Booij J, van den Brink W. The nicotinic acetylcholine receptor partial
agonist varenicline and the treatment of drug dependence: a review. Eur
D

Neuropsychopharmacol 2010;20:69-79.
Crunelle CL, Schulz S, de Bruin K, Miller ML, van den Brink W, Booij J. Dose-dependent and
TE

sustained effects of varenicline on dopamine D2/3 receptor availability in rats. Eur


Neuropsychopharmacol 2011;21:205-10.
Dackis CA, Kampman KM, Lynch KG, Pettinati HM, O'Brien CP. A double-blind, placebo-
P

controlled trial of modafinil for cocaine dependence. Neuropsychopharmacology


2005;30:205-11.
CE

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 Abuse
Treat 2012;43:303-12.
Dai X, Thavundayil J, Gianoulakis C. Differences in the peripheral levels of beta-endorphin in
AC

response to alcohol and stress as a function of alcohol dependence and family history
of alcoholism. Alcohol Clin Exp Res 2005;29:1965-75.
Darke S, Farrell M. Which medications are suitable for agonist drug maintenance? Addiction
2015;In Press.
Daskalakis ZJ, Levinson AJ, Fitzgerald PB. Repetitive transcranial magnetic stimulation for
major depressive disorder: a review. Can J Psychiatry 2008;53:555-66.
Davies DL, Bortolato M, Finn DA, Ramaker MJ, Barak S, Ron D, et al. Recent advances in the
discovery and preclinical testing of novel compounds for the prevention and/or
treatment of alcohol use disorders. Alcohol Clin Exp Res 2013;37:8-15.
de Haan S, Rietveld E, Stokhof M, Denys D. Effects of Deep Brain Stimulation on the Lived
Experience of Obsessive-Compulsive Disorder Patients: In-Depth Interviews with 18
Patients. PLoS One 2015;10:e0135524.
Degenhardt L, Bucello C, Mathers B, Briegleb C, Ali H, Hickman M, et al. Mortality among
regular or dependent users of heroin and other opioids: a systematic review and
meta-analysis of cohort studies. Addiction 2011;106:32-51.
Degenhardt L, Charlson F, Mathers B, Hall WD, Flaxman AD, Johns N, et al. The global
epidemiology and burden of opioid dependence: results from the global burden of
disease 2010 study. Addiction 2014;109:1320-33.
Degenhardt L, Larney S, Randall D, Burns L, Hall W. Causes of death in a cohort treated for
opioid dependence between 1985 and 2005. Addiction 2013.

29
ACCEPTED MANUSCRIPT

Dixon R, Gentile J, Hsu HB, Hsiao J, Howes J, Garg D, et al. Nalmefene: safety and kinetics
after single and multiple oral doses of a new opioid antagonist. J Clin Pharmacol
1987;27:233-9.
Donoghue K, Elzerbi C, Saunders R, Whittington C, Pilling S, Drummond C. The efficacy of
acamprosate and naltrexone in the treatment of alcohol dependence, Europe versus
the rest of the world: a meta-analysis. Addiction 2015;110:920-30.
Drobes DJ, Anton RF, Thomas SE, Voronin K. A clinical laboratory paradigm for evaluating

T
medication effects on alcohol consumption: naltrexone and nalmefene.
Neuropsychopharmacology 2003;28:755-64.

IP
Drobes DJ, Anton RF, Thomas SE, Voronin K. Effects of naltrexone and nalmefene on
subjective response to alcohol among non-treatment-seeking alcoholics and social
drinkers. Alcohol Clin Exp Res 2004;28:1362-70.

R
Dunlop K, Woodside B, Olmsted M, Colton P, Giacobbe P, Downar J. Reductions in Cortico-
Striatal Hyperconnectivity Accompany Successful Treatment of Obsessive-Compulsive

SC
Disorder with Dorsomedial Prefrontal rTMS. Neuropsychopharmacology 2015.
Dursteler KM. The Brain-Behavioral Connection in Substance Use Disorders and Effects
Associated with Injectable Opioid Prescription. Munich: Herbert Utz Verlag; 2015.

NU
Dursteler KM, Berger EM, Strasser J, Caflisch C, Mutschler J, Herdener M, et al. Clinical
potential of methylphenidate in the treatment of cocaine addiction: a review of the
current evidence. Substance abuse and rehabilitation 2015;6:61-74.
Dutra L, Stathopoulou G, Basden SL, Leyro TM, Powers MB, Otto MW. A meta-analytic review
MA
of psychosocial interventions for substance use disorders. Am J Psychiatry
2008;165:179-87.
Edwards S, Kenna GA, Swift RM, Leggio L. Current and promising pharmacotherapies, and
novel research target areas in the treatment of alcohol dependence: a review. Curr
Pharm Des 2011;17:1323-32.
D

Ehrenreich H, Krampe H. Does disulfiram have a role in alcoholism treatment today? Not to
forget about disulfiram's psychological effects. Addiction 2004;99:26-7; author reply
TE

7-8.
EMA. Selincro (nalmefene): Summary of Product Characteristics. London: European
Medicines Agency; 2014.
P

EMCDDA. European Drug Report 2014: Trends and developments. Lisbon: Publications Office
of the European Union; 2014.
CE

EMCDDA. Mortality among drug users in Europe: new and old challenges for public health.
Luxembourg: Publications Office of the European Union; 2015.
Emmerson PJ, Liu MR, Woods JH, Medzihradsky F. Binding affinity and selectivity of opioids
at mu, delta and kappa receptors in monkey brain membranes. J Pharmacol Exp Ther
AC

1994;271:1630-7.
Emsley E, Lees R, Lingford-Hughes A, Nutt D. A review of stress and endogenous opioid
interaction in alcohol addiction. J Neurol Neurosurg Psychiatry 2013;84:e1.
Epstein T, Patsopoulos NA, Weiser M. Immediate-release methylphenidate for attention
deficit hyperactivity disorder (ADHD) in adults. Cochrane Database Syst Rev
2014;9:CD005041.
Erwin BL, Slaton RM. Varenicline in the Treatment of Alcohol Use Disorders. Ann
Pharmacother 2014;48:1445-55.
Faiman MD, Kaul S, Latif SA, Williams TD, Lunte CE. S-(N, N-diethylcarbamoyl)glutathione
(carbamathione), a disulfiram metabolite and its effect on nucleus accumbens and
prefrontal cortex dopamine, GABA, and glutamate: a microdialysis study.
Neuropharmacology 2013;75:95-105.
Feil J, Zangen A. Brain stimulation in the study and treatment of addiction. Neuroscience and
biobehavioral reviews 2010;34:559-74.
Ferri M, Davoli M, Perucci CA. Heroin maintenance for chronic heroin-dependent individuals.
Cochrane Database Syst Rev 2011:CD003410.
Ferri M, Minozzi S, Bo A, Amato L. Slow-release oral morphine as maintenance therapy for
opioid dependence. Cochrane Database Syst Rev 2013;6:CD009879.
Fitzgerald PB, Fountain S, Daskalakis ZJ. A comprehensive review of the effects of rTMS on
motor cortical excitability and inhibition. Clin Neurophysiol 2006;117:2584-96.

30
ACCEPTED MANUSCRIPT

Fitzgerald PB, Maller JJ, Hoy K, Farzan F, Daskalakis ZJ. GABA and cortical inhibition in
motor and non-motor regions using combined TMS-EEG: a time analysis. Clin
Neurophysiol 2009;120:1706-10.
Franck J, Jayaram-Lindstrom N. Pharmacotherapy for alcohol dependence: status of current
treatments. Curr Opin Neurobiol 2013;23:692-9.
French JA, Kanner AM, Bautista J, Abou-Khalil B, Browne T, Harden CL, et al. Efficacy and
tolerability of the new antiepileptic drugs II: treatment of refractory epilepsy: report

T
of the Therapeutics and Technology Assessment Subcommittee and Quality Standards
Subcommittee of the American Academy of Neurology and the American Epilepsy

IP
Society. Neurology 2004;62:1261-73.
Froehlich JC, Hausauer BJ, Rasmussen DD. Combining naltrexone and prazosin in a single
oral medication decreases alcohol drinking more effectively than does either drug

R
alone. Alcohol Clin Exp Res 2013;37:1763-70.
Gahr M, Freudenmann RW, Hiemke C, Kolle MA, Schonfeldt-Lecuona C. Abuse of

SC
methylphenidate in Germany: data from spontaneous reports of adverse drug
reactions. Psychiatry Res 2014;215:252-4.
Garbutt JC, Kampov-Polevoy AB, Gallop R, Kalka-Juhl L, Flannery BA. Efficacy and safety of

NU
baclofen for alcohol dependence: a randomized, double-blind, placebo-controlled trial.
Alcohol Clin Exp Res 2010;34:1849-57.
Garcia-Portilla MP, Bobes-Bascaran MT, Bascaran MT, Saiz PA, Bobes J. Long term outcomes
of pharmacological treatments for opioid dependence: does methadone still lead the
MA
pack? Br J Clin Pharmacol 2014;77:272-84.
Gelernter J, Gueorguieva R, Kranzler HR, Zhang H, Cramer J, Rosenheck R, et al. Opioid
receptor gene (OPRM1, OPRK1, and OPRD1) variants and response to naltrexone
treatment for alcohol dependence: results from the VA Cooperative Study. Alcohol
Clin Exp Res 2007;31:555-63.
D

Gianoulakis C. Endogenous opioids and addiction to alcohol and other drugs of abuse. Curr
Top Med Chem 2004;4:39-50.
TE

Giuliano C, Goodlett CR, Economidou D, Garcia-Pardo MP, Belin D, Robbins TW, et al. The
Novel mu-Opioid Receptor Antagonist GSK1521498 Decreases Both Alcohol Seeking
and Drinking: Evidence from a New Preclinical Model of Alcohol Seeking.
P

Neuropsychopharmacology 2015.
Gorelick DA, Zangen A, George MS. Transcranial magnetic stimulation in the treatment of
CE

substance addiction. Ann N Y Acad Sci 2014;1327:79-93.


Gowing LR, Ali RL, Allsop S, Marsden J, Turf EE, West R, et al. Global statistics on addictive
behaviours: 2014 status report. Addiction 2015;110:904-19.
Grammer GG, Kuhle AR, Clark CC, Dretsch MN, Williams KA, Cole JT. Severity of Depression
AC

Predicts Remission Rates Using Transcranial Magnetic Stimulation. Front Psychiatry


2015;6:114.
Grant BF, Goldstein RB, Saha TD, Chou SP, Jung J, Zhang H, et al. Epidemiology of DSM-5
Alcohol Use Disorder: Results From the National Epidemiologic Survey on Alcohol and
Related Conditions III. JAMA psychiatry 2015;72:757-66.
Grant BF, Stinson FS, Dawson DA, Chou SP, Ruan WJ, Pickering RP. Co-occurrence of 12-
month alcohol and drug use disorders and personality disorders in the United States:
results from the National Epidemiologic Survey on Alcohol and Related Conditions.
Arch Gen Psychiatry 2004;61:361-8.
Grassi MC, Cioce AM, Giudici FD, Antonilli L, Nencini P. Short-term efficacy of Disulfiram or
Naltrexone in reducing positive urinalysis for both cocaine and cocaethylene in
cocaine abusers: a pilot study. Pharmacological research : the official journal of the
Italian Pharmacological Society 2007;55:117-21.
Greenberg BD, Askland KD, Carpenter LL. The evolution of deep brain stimulation for
neuropsychiatric disorders. Frontiers in bioscience : a journal and virtual library
2008;13:4638-48.
Groshkova T, Metrebian N, Hallam C, Charles V, Martin A, Forzisi L, et al. Treatment
expectations and satisfaction of treatment-refractory opioid-dependent patients in
RIOTT, the Randomised Injectable Opiate Treatment Trial, the UK's first supervised
injectable maintenance clinics. Drug Alcohol Rev 2013;32:566-73.

31
ACCEPTED MANUSCRIPT

Gual A, He Y, Torup L, van den Brink W, Mann K. A randomised, double-blind, placebo-


controlled, efficacy study of nalmefene, as-needed use, in patients with alcohol
dependence. Eur Neuropsychopharmacol 2013;23:1432-42.
Gueorguieva R, Wu R, Donovan D, Rounsaville BJ, Couper D, Krystal JH, et al. Baseline
trajectories of drinking moderate acamprosate and naltrexone effects in the COMBINE
study. Alcohol Clin Exp Res 2011;35:523-31.
Guglielmo R, Martinotti G, Quatrale M, Ioime L, Kadilli I, Di Nicola M, et al. Topiramate in

T
Alcohol Use Disorders: Review and Update. CNS Drugs 2015;29:383-95.
Hammig R, Kohler W, Bonorden-Kleij K, Weber B, Lebentrau K, Berthel T, et al. Safety and

IP
tolerability of slow-release oral morphine versus methadone in the treatment of
opioid dependence. J Subst Abuse Treat 2014;47:275-81.
Harel-Fisch Y, Zadok Y, Dejalovsky A. Trend of substance use among Israel youth: Findings

R
from the Israel-HBSC survey 1994–2011. Jerusalem: Israel Anti-Drug Authority;
2013.

SC
Hasan A, Guse B, Cordes J, Wolwer W, Winterer G, Gaebel W, et al. Cognitive Effects of
High-Frequency rTMS in Schizophrenia Patients With Predominant Negative
Symptoms: Results From a Multicenter Randomized Sham-Controlled Trial. Schizophr

NU
Bull 2015.
Heilig M, Egli M. Pharmacological treatment of alcohol dependence: target symptoms and
target mechanisms. Pharmacol Ther 2006;111:855-76.
Heilig M, Koob GF. A key role for corticotropin-releasing factor in alcohol dependence. Trends
MA
Neurosci 2007;30:399-406.
Heinz A, Reimold M, Wrase J, Hermann D, Croissant B, Mundle G, et al. Correlation of stable
elevations in striatal mu-opioid receptor availability in detoxified alcoholic patients
with alcohol craving: a positron emission tomography study using carbon 11-labeled
carfentanil. Arch Gen Psychiatry 2005;62:57-64.
D

Herremans SC, Van Schuerbeek P, De Raedt R, Matthys F, Buyl R, De Mey J, et al. The
Impact of Accelerated Right Prefrontal High-Frequency Repetitive Transcranial
TE

Magnetic Stimulation (rTMS) on Cue-Reactivity: An fMRI Study on Craving in Recently


Detoxified Alcohol-Dependent Patients. PLoS One 2015;10:e0136182.
Higuchi S. Efficacy of acamprosate for the treatment of alcohol dependence long after
P

recovery from withdrawal syndrome: a randomized, double-blind, placebo-controlled


study conducted in Japan (Sunrise Study). J Clin Psychiatry 2015;76:181-8.
CE

Hulka LM, Scheidegger M, Vonmoos M, Preller KH, Baumgartner MR, Herdener M, et al.
Glutamatergic and neurometabolic alterations in chronic cocaine users measured with
H-magnetic resonance spectroscopy. Addict Biol 2014.
Humphreys KL, Eng T, Lee SS. Stimulant medication and substance use outcomes: a meta-
AC

analysis. JAMA psychiatry 2013;70:740-9.


Imbert B, Alvarez JC, Simon N. Anticraving Effect of Baclofen in Alcohol-Dependent Patients.
Alcohol Clin Exp Res 2015;39:1602-8.
Imtiaz S, Shield KD, Fischer B, Rehm J. Harms of prescription opioid use in the United
States. Subst Abuse Treat Prev Policy 2014;9:43.
Ingman K, Hagelberg N, Aalto S, Nagren K, Juhakoski A, Karhuvaara S, et al. Prolonged
central mu-opioid receptor occupancy after single and repeated nalmefene dosing.
Neuropsychopharmacology 2005;30:2245-53.
Jarosz J, Miernik K, Wachal M, Walczak J, Krumpl G. Naltrexone (50 mg) plus psychotherapy
in alcohol-dependent patients: a meta-analysis of randomized controlled trials. Am J
Drug Alcohol Abuse 2013;39:144-60.
Johnson BA. Recent advances in the development of treatments for alcohol and cocaine
dependence: focus on topiramate and other modulators of GABA or glutamate
function. CNS Drugs 2005;19:873-96.
Johnson BA. Update on neuropharmacological treatments for alcoholism: scientific basis and
clinical findings. Biochem Pharmacol 2008;75:34-56.
Johnson BA, Ait-Daoud N. Topiramate in the new generation of drugs: efficacy in the
treatment of alcoholic patients. Curr Pharm Des 2010;16:2103-12.

32
ACCEPTED MANUSCRIPT

Johnson BA, Ait-Daoud N, Akhtar FZ, Ma JZ. Oral topiramate reduces the consequences of
drinking and improves the quality of life of alcohol-dependent individuals: a
randomized controlled trial. Arch Gen Psychiatry 2004;61:905-12.
Johnson BA, Ait-Daoud N, Seneviratne C, Roache JD, Javors MA, Wang XQ, et al.
Pharmacogenetic approach at the serotonin transporter gene as a method of reducing
the severity of alcohol drinking. Am J Psychiatry 2011;168:265-75.
Johnson BA, Roache JD, Javors MA, DiClemente CC, Cloninger CR, Prihoda TJ, et al.

T
Ondansetron for reduction of drinking among biologically predisposed alcoholic
patients: A randomized controlled trial. Jama 2000;284:963-71.

IP
Jonas DE, Amick HR, Feltner C, Bobashev G, Thomas K, Wines R, et al. Pharmacotherapy for
adults with alcohol use disorders in outpatient settings: a systematic review and
meta-analysis. Jama 2014;311:1889-900.

R
June HL, Grey C, Warren-Reese C, Durr LF, Ricks-Cord A, Johnson A, et al. The opioid
receptor antagonist nalmefene reduces responding maintained by ethanol

SC
presentation: preclinical studies in ethanol-preferring and outbred Wistar rats. Alcohol
Clin Exp Res 1998;22:2174-85.
Kalivas PW, Volkow ND. New medications for drug addiction hiding in glutamatergic

NU
neuroplasticity. Mol Psychiatry 2011;16:974-86.
Karhuvaara S, Simojoki K, Virta A, Rosberg M, Loyttyniemi E, Nurminen T, et al. Targeted
nalmefene with simple medical management in the treatment of heavy drinkers: a
randomized double-blind placebo-controlled multicenter study. Alcohol Clin Exp Res
MA
2007;31:1179-87.
Kate N, Grover S, Ghormode D. Dependence on supratherapeutic doses of modafinil: a case
report. The primary care companion for CNS disorders 2012;14.
Kaye S, Darke S. Injecting and non-injecting cocaine use in Sydney, Australia: physical and
psychological morbidity. Drug Alcohol Rev 2004;23:391-8.
D

Keating GM. Sodium oxybate: a review of its use in alcohol withdrawal syndrome and in the
maintenance of abstinence in alcohol dependence. Clin Drug Investig 2014;34:63-80.
TE

Kern DS, Kumar R. Deep brain stimulation. The neurologist 2007;13:237-52.


Kessler RC, Avenevoli S, Costello EJ, Georgiades K, Green JG, Gruber MJ, et al. Prevalence,
persistence, and sociodemographic correlates of DSM-IV disorders in the National
P

Comorbidity Survey Replication Adolescent Supplement. Arch Gen Psychiatry


2012;69:372-80.
CE

Kessler RC, Chiu WT, Demler O, Merikangas KR, Walters EE. Prevalence, severity, and
comorbidity of 12-month DSM-IV disorders in the National Comorbidity Survey
Replication. Arch Gen Psychiatry 2005;62:617-27.
Khantzian EJ. An extreme case of cocaine dependence and marked improvement with
AC

methylphenidate treatment. Am J Psychiatry 1983;140:784-5.


Kim SG, Kim CM, Choi SW, Jae YM, Lee HG, Son BK, et al. A micro opioid receptor gene
polymorphism (A118G) and naltrexone treatment response in adherent Korean
alcohol-dependent patients. Psychopharmacology (Berl) 2009;201:611-8.
Kleber HD, Weiss RD, Anton RF, Jr., George TP, Greenfield SF, Kosten TR, et al. Treatment
of patients with substance use disorders, second edition. American Psychiatric
Association. Am J Psychiatry 2007;164:5-123.
Knapp CM, Tozier L, Pak A, Ciraulo DA, Kornetsky C. Deep brain stimulation of the nucleus
accumbens reduces ethanol consumption in rats. Pharmacol Biochem Behav
2009;92:474-9.
Knoch D, Gianotti LR, Pascual-Leone A, Treyer V, Regard M, Hohmann M, et al. Disruption of
right prefrontal cortex by low-frequency repetitive transcranial magnetic stimulation
induces risk-taking behavior. J Neurosci 2006;26:6469-72.
Knudsen HK, Roman PM. Dissemination, adoption, and implementation of acamprosate for
treating alcohol use disorders. J Stud Alcohol Drugs 2014;75:467-75.
Kollins SH, MacDonald EK, Rush CR. Assessing the abuse potential of methylphenidate in
nonhuman and human subjects: a review. Pharmacol Biochem Behav 2001;68:611-
27.
Koob G, Arends M, Le Moal M. Drugs, Addiction, and the Brain. Amsterdam, Boston,
Heidelberg: Academic Press; 2014.

33
ACCEPTED MANUSCRIPT

Koob GF, Roberts AJ, Kieffer BL, Heyser CJ, Katner SN, Ciccocioppo R, et al. Animal models
of motivation for drinking in rodents with a focus on opioid receptor
neuropharmacology. Recent Dev Alcohol 2003;16:263-81.
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;140:42-7.
Kosten TR, Wu G, Huang W, Harding MJ, Hamon SC, Lappalainen J, et al. Pharmacogenetic

T
randomized trial for cocaine abuse: disulfiram and dopamine beta-hydroxylase. Biol
Psychiatry 2013;73:219-24.

IP
Kranzler HR, Armeli S, Covault J, Tennen H. Variation in OPRM1 moderates the effect of
desire to drink on subsequent drinking and its attenuation by naltrexone treatment.
Addict Biol 2013;18:193-201.

R
Kranzler HR, Armeli S, Feinn R, Tennen H, Gelernter J, Covault J. GRIK1 genotype
moderates topiramate's effects on daily drinking level, expectations of alcohol's

SC
positive effects and desire to drink. Int J Neuropsychopharmacol 2014a;17:1549-56.
Kranzler HR, Covault J, Feinn R, Armeli S, Tennen H, Arias AJ, et al. Topiramate treatment
for heavy drinkers: moderation by a GRIK1 polymorphism. Am J Psychiatry

NU
2014b;171:445-52.
Kravitz AV, Tomasi D, LeBlanc KH, Baler R, Volkow ND, Bonci A, et al. Cortico-striatal
circuits: Novel therapeutic targets for substance use disorders. Brain Res 2015.
Krishnan R, Chary KV. A rare case modafinil dependence. Journal of pharmacology &
MA
pharmacotherapeutics 2015;6:49-50.
Krupitsky E, Nunes EV, Ling W, Illeperuma A, Gastfriend DR, Silverman BL. Injectable
extended-release naltrexone for opioid dependence: a double-blind, placebo-
controlled, multicentre randomised trial. Lancet 2011;377:1506-13.
Kuhn J, Bauer R, Pohl S, Lenartz D, Huff W, Kim EH, et al. Observations on unaided smoking
D

cessation after deep brain stimulation of the nucleus accumbens. Eur Addict Res
2009;15:196-201.
TE

Kuhn J, Bodatsch M, Sturm V, Lenartz D, Klosterkotter J, Uhlhaas PJ, et al. [Deep brain
stimulation in schizophrenia]. Fortschr Neurol Psychiatr 2011a;79:632-41.
Kuhn J, Grundler TO, Bauer R, Huff W, Fischer AG, Lenartz D, et al. Successful deep brain
P

stimulation of the nucleus accumbens in severe alcohol dependence is associated with


changed performance monitoring. Addict Biol 2011b;16:620-3.
CE

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.
Leonard BE, McCartan D, White J, King DJ. Methylphenidate: a review of its
AC

neuropharmacological, neuropsychological and adverse clinical effects. Human


psychopharmacology 2004;19:151-80.
Levin FR, Evans SM, Brooks DJ, Garawi F. Treatment of cocaine dependent treatment
seekers with adult ADHD: double-blind comparison of methylphenidate and placebo.
Drug Alcohol Depend 2007;87:20-9.
Levkovitz Y, Roth Y, Harel EV, Braw Y, Sheer A, Zangen A. A randomized controlled
feasibility and safety study of deep transcranial magnetic stimulation. Clin
Neurophysiol 2007;118:2730-44.
Likhitsathian S, Uttawichai K, Booncharoen H, Wittayanookulluk A, Angkurawaranon C,
Srisurapanont M. Topiramate treatment for alcoholic outpatients recently receiving
residential treatment programs: a 12-week, randomized, placebo-controlled trial.
Drug Alcohol Depend 2013;133:440-6.
Ling W, Casadonte P, Bigelow G, Kampman KM, Patkar A, Bailey GL, et al. Buprenorphine
implants for treatment of opioid dependence: a randomized controlled trial. Jama
2010;304:1576-83.
Lingford-Hughes AR, Welch S, Peters L, Nutt DJ. BAP updated guidelines: evidence-based
guidelines for the pharmacological management of substance abuse, harmful use,
addiction and comorbidity: recommendations from BAP. J Psychopharmacol
2012;26:899-952.

34
ACCEPTED MANUSCRIPT

Litten RZ, Ryan M, Falk D, Fertig J. Alcohol medications development: advantages and
caveats of government/academia collaborating with the pharmaceutical industry.
Alcohol Clin Exp Res 2014;38:1196-9.
Litten RZ, Ryan ML, Fertig JB, Falk DE, Johnson B, Dunn KE, et al. A double-blind, placebo-
controlled trial assessing the efficacy of varenicline tartrate for alcohol dependence. J
Addict Med 2013;7:277-86.
Littleton J, Zieglgansberger W. Pharmacological mechanisms of naltrexone and acamprosate

T
in the prevention of relapse in alcohol dependence. Am J Addict 2003;12 Suppl 1:S3-
11.

IP
Liu HY, 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;13:40-6.

R
Lobmaier PP, Kunoe N, Gossop M, Waal H. Naltrexone depot formulations for opioid and
alcohol dependence: a systematic review. CNS Neurosci Ther 2011;17:629-36.

SC
Luigjes J, van den Brink W, Feenstra M, van den Munckhof P, Schuurman PR, Schippers R, et
al. Deep brain stimulation in addiction: a review of potential brain targets. Mol
Psychiatry 2012;17:572-83.

NU
Luigjes J, van den Brink W, Schuurman PR, Kuhn J, Denys D. Is deep brain stimulation a
treatment option for addiction? Addiction 2015;110:547-8.
Madlung-Kratzer E, Spitzer B, Brosch R, Dunkel D, Haring C. A double-blind, randomized,
parallel group study to compare the efficacy, safety and tolerability of slow-release
MA
oral morphine versus methadone in opioid-dependent in-patients willing to undergo
detoxification. Addiction 2009;104:1549-57.
Madry MM, Steuer AE, Vonmoos M, Quednow BB, Baumgartner MR, Kraemer T.
Retrospective monitoring of long-term recreational and dependent cocaine use in
toenail clippings/scrapings as an alternative to hair. Bioanalysis 2014;6:3183-96.
D

Maier LJ, Wunderli MD, Vonmoos M, Rommelt AT, Baumgartner MR, Seifritz E, et al.
Pharmacological Cognitive Enhancement in Healthy Individuals: A Compensation for
TE

Cognitive Deficits or a Question of Personality? PLoS One 2015;10:e0129805.


Maisel NC, Blodgett JC, Wilbourne PL, Humphreys K, Finney JW. Meta-analysis of naltrexone
and acamprosate for treating alcohol use disorders: when are these medications most
P

helpful? Addiction 2013;108:275-93.


Mammen K, Bell J. The clinical efficacy and abuse potential of combination buprenorphine-
CE

naloxone in the treatment of opioid dependence. Expert Opin Pharmacother


2009;10:2537-44.
Mann K, Bladstrom A, Torup L, Gual A, van den Brink W. Extending the treatment options in
alcohol dependence: a randomized controlled study of as-needed nalmefene. Biol
AC

Psychiatry 2013a;73:706-13.
Mann K, Lemenager T, Hoffmann S, Reinhard I, Hermann D, Batra A, et al. Results of a
double-blind, placebo-controlled pharmacotherapy trial in alcoholism conducted in
Germany and comparison with the US COMBINE study. Addict Biol 2013b;18:937-46.
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; discussion E.
Mariani JJ, Hart CL. Psychosis associated with modafinil and shift work. Am J Psychiatry
2005;162:1983.
Marinelli PW, Bai L, Quirion R, Gianoulakis C. A microdialysis profile of Met-enkephalin
release in the rat nucleus accumbens following alcohol administration. Alcohol Clin
Exp Res 2005;29:1821-8.
Marinelli PW, Lam M, Bai L, Quirion R, Gianoulakis C. A microdialysis profile of dynorphin
A(1-8) release in the rat nucleus accumbens following alcohol administration. Alcohol
Clin Exp Res 2006;30:982-90.
Marinelli PW, Quirion R, Gianoulakis C. An in vivo profile of beta-endorphin release in the
arcuate nucleus and nucleus accumbens following exposure to stress or alcohol.
Neuroscience 2004;127:777-84.
Marzuk PM, Tardiff K, Leon AC, Hirsch CS, Portera L, Iqbal MI, et al. Ambient temperature
and mortality from unintentional cocaine overdose. Jama 1998;279:1795-800.

35
ACCEPTED MANUSCRIPT

Marzuk PM, Tardiff K, Leon AC, Stajic M, Morgan EB, Mann JJ. Prevalence of cocaine use
among residents of New York City who committed suicide during a one-year period.
Am J Psychiatry 1992;149:371-5.
Mason BJ, Ritvo EC, Morgan RO, Salvato FR, Goldberg G, Welch B, et al. A double-blind,
placebo-controlled pilot study to evaluate the efficacy and safety of oral nalmefene
HCl for alcohol dependence. Alcohol Clin Exp Res 1994;18:1162-7.
Mason BJ, Salvato FR, Williams LD, Ritvo EC, Cutler RB. A double-blind, placebo-controlled

T
study of oral nalmefene for alcohol dependence. Arch Gen Psychiatry 1999;56:719-
24.

IP
Mattick RP, Breen C, Kimber J, Davoli M. Methadone maintenance therapy versus no opioid
replacement therapy for opioid dependence. Cochrane Database Syst Rev
2009a:CD002209.

R
Mattick RP, Breen C, Kimber J, Davoli M. Methadone maintenance therapy versus no opioid
replacement therapy for opioid dependence. Cochrane Database Syst Rev

SC
2009b:CD002209.
Mattick RP, Breen C, Kimber J, Davoli M. Buprenorphine maintenance versus placebo or
methadone maintenance for opioid dependence. The Cochrane database of

NU
systematic reviews 2014;2:CD002207.
McCance-Katz EF, Kosten TR, Jatlow P. Disulfiram effects on acute cocaine administration.
Drug Alcohol Depend 1998;52:27-39.
Mishra BR, Praharaj SK, Katshu MZ, Sarkar S, Nizamie SH. Comparison of anticraving
MA
efficacy of right and left repetitive transcranial magnetic stimulation in alcohol
dependence: a randomized double-blind study. J Neuropsychiatry Clin Neurosci
2015;27:e54-9.
Mitchell JM, Teague CH, Kayser AS, Bartlett SE, Fields HL. Varenicline decreases alcohol
consumption in heavy-drinking smokers. Psychopharmacology (Berl) 2012;223:299-
D

306.
Morley KC, Baillie A, Leung S, Addolorato G, Leggio L, Haber PS. Baclofen for the treatment
TE

of alcohol dependence and possible role of comorbid anxiety. Alcohol Alcohol 2014.
Muller CA, Geisel O, Banas R, Heinz A. Current pharmacological treatment approaches for
alcohol dependence. Expert Opin Pharmacother 2014;15:471-81.
P

Muller CA, Geisel O, Pelz P, Higl V, Kruger J, Stickel A, et al. High-dose baclofen for the
treatment of alcohol dependence (BACLAD study): A randomized, placebo-controlled
CE

trial. Eur Neuropsychopharmacol 2015;25:1167-77.


Muller UJ, Sturm V, Voges J, Heinze HJ, Galazky I, Heldmann M, et al. Successful treatment
of chronic resistant alcoholism by deep brain stimulation of nucleus accumbens: first
experience with three cases. Pharmacopsychiatry 2009;42:288-91.
AC

Mutschler J, Buhler M, Diehl A, Mann K, Kiefer F. Disulfiram, an old drug with new potential
in the treatment of pathological gambling? Medical hypotheses 2010;74:209-10.
Mutschler J, Diehl A, Kiefer F. Pronounced paranoia as a result of cocaine-disulfiram
interaction: case report and mode of action. J Clin Psychopharmacol 2009;29:99-101.
Mutschler J, Dirican G, Funke S, Obermann C, Grosshans M, Mann K, et al. Experienced
acetaldehyde reaction does not improve treatment response in outpatients treated
with supervised disulfiram. Clin Neuropharmacol 2011;34:161-5.
Mutschler J, Kiefer F. [Mechanism of action of disulfiram and treatment optimization in
prevention of recurrent alcoholism]. Praxis 2013;102:139-46.
Myrick H, Malcolm R, Brady KT. Gabapentin treatment of alcohol withdrawal. Am J Psychiatry
1998;155:1632.
Narita M, Funada M, Suzuki T. Regulations of opioid dependence by opioid receptor types.
Pharmacol Ther 2001;89:1-15.
New South Wales Department of Health. Opioid treatment program: Clinical guidelines for
methadone and buprenorphine treatment. Sydney: NSW Government; 2006.
Niciu MJ, Arias AJ. Targeted opioid receptor antagonists in the treatment of alcohol use
disorders. CNS Drugs 2013;27:777-87.
Nocente R, Vitali M, Balducci G, Enea D, Kranzler HR, Ceccanti M. Varenicline and neuronal
nicotinic acetylcholine receptors: a new approach to the treatment of co-occurring
alcohol and nicotine addiction? Am J Addict 2013;22:453-9.

36
ACCEPTED MANUSCRIPT

Nordt C, Stohler R. Low-threshold methadone treatment, heroin price, police activity and
incidence of heroin use: the Zurich experience. Int J Drug Policy 2009;20:497-501.
Noronha ABC, Cui C, Harris RA, Crabbe JC. Neurobiology of Alcohol Dependence. London,
Waltham, San Diego: Academic Press; 2014.
Nuijten M, Blanken P, van den Brink W, Hendriks V. Modafinil in the treatment of crack-
cocaine dependence in the Netherlands: Results of an open-label randomised
controlled feasibility trial. J Psychopharmacol 2015;29:678-87.

T
Nutt D, Nestor L. Addiction. Kettering: Oxford University Press; 2013.
Nutt DJ. The role of the opioid system in alcohol dependence. J Psychopharmacol 2014;28:8-

IP
22.
O'Malley SS, Corbin WR, Leeman RF, DeMartini KS, Fucito LM, Ikomi J, et al. Reduction of
alcohol drinking in young adults by naltrexone: a double-blind, placebo-controlled,

R
randomized clinical trial of efficacy and safety. J Clin Psychiatry 2015;76:e207-13.
Olds J, Milner P. Positive reinforcement produced by electrical stimulation of septal area and

SC
other regions of rat brain. Journal of comparative and physiological psychology
1954;47:419-27.
Oliveto A, Poling J, Mancino MJ, Feldman Z, Cubells JF, Pruzinsky R, et al. Randomized,

NU
double blind, placebo-controlled trial of disulfiram for the treatment of cocaine
dependence in methadone-stabilized patients. Drug Alcohol Depend 2011;113:184-
91.
Oroszi G, Anton RF, O'Malley S, Swift R, Pettinati H, Couper D, et al. OPRM1 Asn40Asp
MA
predicts response to naltrexone treatment: a haplotype-based approach. Alcohol Clin
Exp Res 2009;33:383-93.
Oslin DW, Leong SH, Lynch KG, Berrettini W, O'Brien CP, Gordon AJ, et al. Naltrexone vs
Placebo for the Treatment of Alcohol Dependence: A Randomized Clinical Trial. JAMA
psychiatry 2015;72:430-7.
D

Pani PP, Trogu E, Pacini M, Maremmani I. Anticonvulsants for alcohol dependence. Cochrane
Database Syst Rev 2014;2:CD008544.
TE

Parran TV, Jr., Jasinski DR. Intravenous methylphenidate abuse. Prototype for prescription
drug abuse. Arch Intern Med 1991;151:781-3.
Paslakis G, Kiefer F, Diehl A, Alm B, Sobanski E. [Methylphenidate. Therapy option for adults
P

with ADHD and comorbid substance use disorder?]. Nervenarzt 2010;81:277-88.


Peles E, Schreiber S, Adelson M. 15-Year survival and retention of patients in a general
CE

hospital-affiliated methadone maintenance treatment (MMT) center in Israel. Drug


Alcohol Depend 2010;107:141-8.
Peles E, Schreiber S, Linzy S, Domani Y, Adelson M. Differences in methylphenidate abuse
rates among methadone maintenance treatment patients in two clinics. J Subst Abuse
AC

Treat 2015;54:44-9.
Petrakis IL, Carroll KM, Nich C, Gordon LT, McCance-Katz EF, Frankforter T, et al. Disulfiram
treatment for cocaine dependence in methadone-maintained opioid addicts. Addiction
2000;95:219-28.
Pettinati HM, Kampman KM, Lynch KG, Xie H, Dackis C, Rabinowitz AR, et al. A double blind,
placebo-controlled trial that combines disulfiram and naltrexone for treating co-
occurring cocaine and alcohol dependence. Addict Behav 2008;33:651-67.
Pirkola SP, Poikolainen K, Lonnqvist JK. Currently active and remitted alcohol dependence in
a nationwide adult general population--results from the Finnish Health 2000 study.
Alcohol Alcohol 2006;41:315-20.
Plebani JG, Lynch KG, Rennert L, Pettinati HM, O'Brien CP, Kampman KM. Results from a
pilot clinical trial of varenicline for the treatment of alcohol dependence. Drug Alcohol
Depend 2013;133:754-8.
Ponizovsky AM, Rosca P, Aronovich E, Weizman A, Grinshpoon A. Baclofen as add-on to
standard psychosocial treatment for alcohol dependence: a randomized, double-blind,
placebo-controlled trial with 1 year follow-up. J Subst Abuse Treat 2015;52:24-30.
Preti A. New developments in the pharmacotherapy of cocaine abuse. Addict Biol
2007;12:133-51.

37
ACCEPTED MANUSCRIPT

Prikryl R, Ustohal L, Kucerova HP, Kasparek T, Jarkovsky J, Hublova V, et al. Repetitive


transcranial magnetic stimulation reduces cigarette consumption in schizophrenia
patients. Prog Neuropsychopharmacol Biol Psychiatry 2014;49:30-5.
Project MATCH Research Group. Matching alcoholism treatments to client heterogeneity:
treatment main effects and matching effects on drinking during treatment. J Stud
Alcohol 1998;59:631-9.
Protasio MI, da Silva JP, Arias-Carrion O, Nardi AE, Machado S, Cruz MS. Repetitive

T
transcranial magnetic stimulation to treat substance use disorders and compulsive
behavior. CNS Neurol Disord Drug Targets 2015;14:331-40.

IP
Rasmussen DD, Kincaid CL, Froehlich JC. Prazosin + Naltrexone Decreases Alcohol Drinking
More Effectively Than Does Either Drug Alone in P Rats with a Protracted History of
Extensive Voluntary Alcohol Drinking, Dependence, and Multiple Withdrawals. Alcohol

R
Clin Exp Res 2015;39:1832-41.
Rehm J, Room R, van den Brink W, Jacobi F. Alcohol use disorders in EU countries and

SC
Norway: an overview of the epidemiology. Eur Neuropsychopharmacol 2005;15:377-
88.
Ritz MC, Lamb RJ, Goldberg SR, Kuhar MJ. Cocaine receptors on dopamine transporters are

NU
related to self-administration of cocaine. Science 1987;237:1219-23.
Rocha BA. Stimulant and reinforcing effects of cocaine in monoamine transporter knockout
mice. Eur J Pharmacol 2003;479:107-15.
Rogawski MA, Loscher W. The neurobiology of antiepileptic drugs. Nat Rev Neurosci
MA
2004;5:553-64.
Rolland B, Bordet R, Deheul S, Cottencin O. Baclofen for alcohol-dependence: anticraving or
partial substitution? J Clin Psychopharmacol 2013;33:280-1.
Rolland B, Labreuche J, Duhamel A, Deheul S, Gautier S, Auffret M, et al. Baclofen for
alcohol dependence: Relationships between baclofen and alcohol dosing and the
D

occurrence of major sedation. Eur Neuropsychopharmacol 2015.


Rosenthal RN, Ling W, Casadonte P, Vocci F, Bailey GL, Kampman K, et al. Buprenorphine
TE

implants for treatment of opioid dependence: randomized comparison to placebo and


sublingual buprenorphine/naloxone. Addiction 2013;108:2141-9.
Rosner S, Hackl-Herrwerth A, Leucht S, Lehert P, Vecchi S, Soyka M. Acamprosate for
P

alcohol dependence. Cochrane Database Syst Rev 2010a:CD004332.


Rosner S, Hackl-Herrwerth A, Leucht S, Vecchi S, Srisurapanont M, Soyka M. Opioid
CE

antagonists for alcohol dependence. Cochrane Database Syst Rev 2010b:CD001867.


Rossi S, Hallett M, Rossini PM, Pascual-Leone A, Safety of TMSCG. Safety, ethical
considerations, and application guidelines for the use of transcranial magnetic
stimulation in clinical practice and research. Clin Neurophysiol 2009;120:2008-39.
AC

Rouaud T, Lardeux S, Panayotis N, Paleressompoulle D, Cador M, Baunez C. Reducing the


desire for cocaine with subthalamic nucleus deep brain stimulation. Proc Natl Acad Sci
U S A 2010;107:1196-200.
Rustembegovic A, Sofic E, Tahirovic I, Kundurovic Z. A study of gabapentin in the treatment
of tonic-clonic seizures of alcohol withdrawal syndrome. Med Arh 2004;58:5-6.
Schacht JP, Anton RF, Voronin KE, Randall PK, Li X, Henderson S, et al. Interacting effects of
naltrexone and OPRM1 and DAT1 variation on the neural response to alcohol cues.
Neuropsychopharmacology 2013;38:414-22.
Schottenfeld RS, Chawarski MC, Cubells JF, George TP, Lappalainen J, Kosten TR.
Randomized clinical trial of disulfiram for cocaine dependence or abuse during
buprenorphine treatment. Drug Alcohol Depend 2014;136:36-42.
Setiawan E, Pihl RO, Benkelfat C, Leyton M. Influence of the OPRM1 A118G polymorphism on
alcohol-induced euphoria, risk for alcoholism and the clinical efficacy of naltrexone.
Pharmacogenomics 2012;13:1161-72.
Shorter D, Domingo CB, Kosten TR. Emerging drugs for the treatment of cocaine use
disorder: a review of neurobiological targets and pharmacotherapy. Expert Opin
Emerg Drugs 2015;20:15-29.
Shorter D, Nielsen DA, Huang W, Harding MJ, Hamon SC, Kosten TR. Pharmacogenetic
randomized trial for cocaine abuse: disulfiram and alpha1A-adrenoceptor gene
variation. Eur Neuropsychopharmacol 2013;23:1401-7.

38
ACCEPTED MANUSCRIPT

Skala K, Caputo F, Mirijello A, Vassallo G, Antonelli M, Ferrulli A, et al. Sodium oxybate in


the treatment of alcohol dependence: from the alcohol withdrawal syndrome to the
alcohol relapse prevention. Expert Opin Pharmacother 2014;15:245-57.
Skinner MD, Lahmek P, Pham H, Aubin HJ. Disulfiram efficacy in the treatment of alcohol
dependence: a meta-analysis. PLoS One 2014;9:e87366.
Sousa AD. Repetitive Transcranial Magnetic Stimulation (rTMS) in the Management of
Alcohol Dependence and other Substance Abuse Disorders - Emerging Data and

T
Clinical Relevance. Basic and clinical neuroscience 2013;4:271-5.
Soyka M. New developments in the management of opioid dependence: focus on sublingual

IP
buprenorphine-naloxone. Subst Abuse Rehabil 2015;6:1-14.
Soyka M, Kranzler HR, van den Brink W, Krystal J, Moller HJ, Kasper S. The World Federation
of Societies of Biological Psychiatry (WFSBP) guidelines for the biological treatment of

R
substance use and related disorders. Part 2: Opioid dependence. World J Biol
Psychiatry 2011a;12:160-87.

SC
Soyka M, Lieb M. Recent Developments in Pharmacotherapy of Alcoholism.
Pharmacopsychiatry 2015;48:123-35.
Soyka M, Rosner S. Emerging drugs to treat alcoholism. Expert Opin Emerg Drugs

NU
2010;15:695-711.
Soyka M, Trader A, Klotsche J, Backmund M, Buhringer G, Rehm J, et al. Six-year mortality
rates of patients in methadone and buprenorphine maintenance therapy: results from
a nationally representative cohort study. J Clin Psychopharmacol 2011b;31:678-80.
MA
Spanagel R, Kiefer F. Drugs for relapse prevention of alcoholism: ten years of progress.
Trends Pharmacol Sci 2008;29:109-15.
Spanagel R, Noori HR, Heilig M. Stress and alcohol interactions: animal studies and clinical
significance. Trends Neurosci 2014a;37:219-27.
Spanagel R, Vengeliene V. New pharmacological treatment strategies for relapse prevention.
D

Curr Top Behav Neurosci 2013;13:583-609.


Spanagel R, Vengeliene V, Jandeleit B, Fischer WN, Grindstaff K, Zhang X, et al.
TE

Acamprosate produces its anti-relapse effects via calcium. Neuropsychopharmacology


2014b;39:783-91.
Spellicy CJ, Kosten TR, Hamon SC, Harding MJ, Nielsen DA. ANKK1 and DRD2
P

pharmacogenetics of disulfiram treatment for cocaine abuse. Pharmacogenetics and


genomics 2013;23:333-40.
CE

Spence D. Bad medicine: nalmefene in alcohol misuse. Bmj 2014;348:g1531.


Stephen JH, Halpern CH, Barrios CJ, Balmuri U, Pisapia JM, Wolf JA, et al. Deep brain
stimulation compared with methadone maintenance for the treatment of heroin
dependence: a threshold and cost-effectiveness analysis. Addiction 2012;107:624-
AC

34.
Stephens MAC, McCaul ME, Wand GS. The potential role of glucocorticoids and the HPA axis
in alcohol dependence. In: Noronha ABC, Changhai CUI, Harris RA, Crabbe JC,
editors. Neurobiology of Alcohol Dependence. London: Academic Press; 2014. p. 429-
50.
Strafella AP, Paus T, Barrett J, Dagher A. Repetitive transcranial magnetic stimulation of the
human prefrontal cortex induces dopamine release in the caudate nucleus. J Neurosci
2001;21:RC157.
Strang J, Groshkova T, Uchtenhagen A, van den Brink W, Haasen C, Schechter MT, et al.
Heroin on trial: systematic review and meta-analysis of randomised trials of
diamorphine-prescribing as treatment for refractory heroin addictiondagger. Br J
Psychiatry 2015;207:5-14.
Strang J, Metrebian N, Lintzeris N, Potts L, Carnwath T, Mayet S, et al. Supervised injectable
heroin or injectable methadone versus optimised oral methadone as treatment for
chronic heroin addicts in England after persistent failure in orthodox treatment
(RIOTT): a randomised trial. Lancet 2010;375:1885-95.
Suh JJ, Pettinati HM, Kampman KM, O'Brien CP. The status of disulfiram: a half of a century
later. J Clin Psychopharmacol 2006;26:290-302.

39
ACCEPTED MANUSCRIPT

Sun Y, Giacobbe P, Tang CW, Barr MS, Rajji T, Kennedy SH, et al. Deep Brain Stimulation
Modulates Gamma Oscillations and Theta-Gamma Coupling in Treatment Resistant
Depression. Brain Stimul 2015.
Swift RM. Naltrexone and nalmefene: any meaningful difference? Biol Psychiatry
2013;73:700-1.
Syed YY, Keating GM. Extended-release intramuscular naltrexone (VIVITROL(R)): a review of
its use in the prevention of relapse to opioid dependence in detoxified patients. CNS

T
Drugs 2013;27:851-61.
Taylor R, Jr., Raffa RB, Pergolizzi JV, Jr. Naltrexone extended-release injection: an option for

IP
the management of opioid abuse. Subst Abuse Rehabil 2011;2:219-26.
United Nations Office on Drugs and Crime. 2006 World Drug Report. Vienna: UNODC; 2006.
Valencia-Alfonso CE, Luigjes J, Smolders R, Cohen MX, Levar N, Mazaheri A, et al. Effective

R
deep brain stimulation in heroin addiction: a case report with complementary
intracranial electroencephalogram. Biol Psychiatry 2012;71:e35-7.

SC
van de Glind G, Konstenius M, Koeter MW, van Emmerik-van Oortmerssen K, Carpentier PJ,
Kaye S, et al. Variability in the prevalence of adult ADHD in treatment seeking
substance use disorder patients: results from an international multi-center study

NU
exploring DSM-IV and DSM-5 criteria. Drug Alcohol Depend 2014;134:158-66.
van den Brink W, Aubin HJ, Bladstrom A, Torup L, Gual A, Mann K. Efficacy of as-needed
nalmefene in alcohol-dependent patients with at least a high drinking risk level:
results from a subgroup analysis of two randomized controlled 6-month studies.
MA
Alcohol Alcohol 2013;48:570-8.
Voges J, Muller U, Bogerts B, Munte T, Heinze HJ. Deep brain stimulation surgery for alcohol
addiction. World Neurosurg 2013;80:S28 e1-31.
Voronin K, Randall P, Myrick H, Anton R. Aripiprazole effects on alcohol consumption and
subjective reports in a clinical laboratory paradigm--possible influence of self-control.
D

Alcohol Clin Exp Res 2008;32:1954-61.


Walker BM, Koob GF. Pharmacological evidence for a motivational role of kappa-opioid
TE

systems in ethanol dependence. Neuropsychopharmacology 2008;33:643-52.


Walker BM, Valdez GR, McLaughlin JP, Bakalkin G. Targeting dynorphin/kappa opioid
receptor systems to treat alcohol abuse and dependence. Alcohol 2012;46:359-70.
P

Walker BM, Zorrilla EP, Koob GF. Systemic kappa-opioid receptor antagonism by nor-
binaltorphimine reduces dependence-induced excessive alcohol self-administration in
CE

rats. Addict Biol 2011;16:116-9.


Watson WP, Robinson E, Little HJ. The novel anticonvulsant, gabapentin, protects against
both convulsant and anxiogenic aspects of the ethanol withdrawal syndrome.
Neuropharmacology 1997;36:1369-75.
AC

Weinshenker D. Cocaine sobers up. Nat Med 2010;16:969-70.


Weinshenker D, Schroeder JP. There and back again: a tale of norepinephrine and drug
addiction. Neuropsychopharmacology 2007;32:1433-51.
Wenzel RG, Schwarz K, Padiyara RS. Topiramate for migraine prevention. Pharmacotherapy
2006;26:375-87.
White HS, Brown SD, Woodhead JH, Skeen GA, Wolf HH. Topiramate modulates GABA-
evoked currents in murine cortical neurons by a nonbenzodiazepine mechanism.
Epilepsia 2000;41 Suppl 1:S17-20.
WHO. The ICD-10 classification of mental and behavioural disorders : clinical descriptions
and diagnostic guidelines. Geneva: World Health Organization; 1992.
WHO. The World Health Report 2004. Geneva: World Health Organization; 2004.
WHO. Global status report on alcohol and health. Geneva, Switzerland: World Health
Organization; 2011.
WHO. Gamma-hydroxybutyric acid (GHB). Critical Review Report. Expert Committee on Drug
Dependence 35th Meeting, Hammamet, Tunisia, 4-8 June 2012. Geneva: World
Health Orgnization; 2012.
Wilens TE, Biederman J, Spencer TJ, Frances RJ. Comorbidity of attention-deficit
hyperactivity and psychoactive substance use disorders. Hospital & community
psychiatry 1994;45:421-3, 35.

40
ACCEPTED MANUSCRIPT

Zhou H, Xu J, Jiang J. Deep brain stimulation of nucleus accumbens on heroin-seeking


behaviors: a case report. Biol Psychiatry 2011;69:e41-2.
Zindel LR, Kranzler HR. Pharmacotherapy of alcohol use disorders: seventy-five years of
progress. J Stud Alcohol Drugs Suppl 2014;75 Suppl 17:79-88.
Zorrilla EP, Heilig M, de Wit H, Shaham Y. Behavioral, biological, and chemical perspectives
on targeting CRF(1) receptor antagonists to treat alcoholism. Drug Alcohol Depend
2013;128:175-86.

T
R IP
SC
NU
MA
D
P TE
CE
AC

41
ACCEPTED MANUSCRIPT

Highlights

- Nalmefene is an as-needed medication for reduction of alcohol intake.


- Depot/implant buprenorphine, i.v. methadone may be options for opioid use disorder.

T
- Neuromodulation techniques such as TMS and DBS are emerging treatment options.
- Disulfiram, methylphenidate, modafinil are being studied for cocaine use disorder.

R IP
SC
NU
MA
D
P TE
CE
AC

42

You might also like