Download as pdf or txt
Download as pdf or txt
You are on page 1of 23

Neurotherapeutics

https://doi.org/10.1007/s13311-022-01229-4

CURRENT PERSPECTIVES

Deep Brain Stimulation for Addictive Disorders—Where Are We Now?


Jason Yuen1,2   · Abbas Z. Kouzani3 · Michael Berk2 · Susannah J. Tye4,5,6,7 · Aaron E. Rusheen1 · Charles D. Blaha1 ·
Kevin E. Bennet1,8 · Kendall H. Lee1,9 · Hojin Shin1 · Jee Hyun Kim2 · Yoonbae Oh1,9

Accepted: 18 March 2022


© The Author(s) 2022

Abstract
In the face of a global epidemic of drug addiction, neglecting to develop new effective therapies will perpetuate the stagger-
ing human and economic costs of substance use. This review aims to summarize and evaluate the preclinical and clinical
studies of deep brain stimulation (DBS) as a novel therapy for refractory addiction, in hopes to engage and inform future
research in this promising novel treatment avenue. An electronic database search (MEDLINE, EMBASE, Cochrane library)
was performed using keywords and predefined inclusion criteria between 1974 and 6/18/2021 (registered on Open Science
Registry). Selected articles were reviewed in full text and key details were summarized and analyzed to understand DBS’
therapeutic potential and possible mechanisms of action. The search yielded 25 animal and 22 human studies. Animal stud-
ies showed that DBS of targets such as nucleus accumbens (NAc), insula, and subthalamic nucleus reduces drug use and
seeking. All human studies were case series/reports (level 4/5 evidence), mostly targeting the NAc with generally positive
outcomes. From the limited evidence in the literature, DBS, particularly of the NAc, appears to be a reasonable last resort
option for refractory addictive disorders. We propose that future research in objective electrophysiological (e.g., local field
potentials) and neurochemical (e.g., extracellular dopamine levels) biomarkers would assist monitoring the progress of treat-
ment and developing a closed-loop DBS system. Preclinical literature also highlighted the prefrontal cortex as a promising
DBS target, which should be explored in human research.

Keywords  Deep brain stimulation · Addiction · Biomarkers · Animal models · Neuromodulation · Neuropsychiatry

Introduction However, the risk of relapse remains high, with some cit-
ing relapse rates as high as 75 to 98% within 1 year of
Addictive substances pose significant socioeconomic treatment [3]. Investigations into new treatment modali-
impact on society and the healthcare system, costing ties are urgently warranted. To develop novel therapies
hundreds of billion dollars annually in the USA alone [1, for addiction, researchers are focusing on the behavioral
2]. Psychological and adjunctive pharmacological treat- and neuroscience aspects in the biopsychosocial model
ments are the current mainstay for addiction therapeutics. (Fig.  1). Deep brain stimulation (DBS) in particular is

4
* Jee Hyun Kim Queensland Brain Institute, The University of Queensland,
drjeehyunkim@gmail.com St Lucia, QLD 4072, Australia
5
* Yoonbae Oh Department of Psychiatry & Psychology, Mayo Clinic,
oh.yoonbae@mayo.edu Rochester, MN 55905, USA
6
1 Department of Psychiatry, University of Minnesota,
Department of Neurologic Surgery, Mayo Clinic, Rochester,
Minneapolis, MN 55455, USA
MN 55905, USA
7
2 Department of Psychiatry, Emory University, Atlanta,
Deakin University, IMPACT, The Institute for Mental
GA 30322, USA
and Physical Health and Clinical Translation, School
8
of Medicine, Geelong VIC 3216, Australia Division of Engineering, Mayo Clinic, Rochester, MN 55905,
3 USA
School of Engineering, Deakin University,
9
Geelong VIC 3216, Australia Department of Biomedical Engineering, Mayo Clinic,
Rochester, MN 55905, USA

13
Vol.:(0123456789)
J. Yuen et al.

Fig. 1  Biopsychosocial model depicting the different dimensions


in consideration of addiction treatment. The stages of the addiction
cycle of behavior are shown (inset) [140]. Interactions between com-
ponents in different domains lead to addictive behaviors. Created with
Biorender.com Fig. 2  A simplified illustration of the neurotransmitters and neuro-
anatomical structures involved in the pathophysiology of addiction
[13, 19, 20]. Other relevant areas are omitted for clarity. The effect of
a neuroscience-based potential treatment for medically dopamine receptors ­D1-R and ­D2-R is shown in inset. ­D1-R mediates
the direct pathway (positive reinforcement) and D ­ 2-R mediates the
refractory addictive disorders, given its diminishing sur- indirect pathway (negative reinforcement). Italic descriptions denote
gical risks. “Medically refractory” cases have no formal changes secondary to chronic drug use. 5-HT, serotonin; AMPAR,
definition, although these are generally regarded when con- α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor;
ventional treatments repeatedly fail [4]. D1/2R, dopamine 1/2 receptors; DA, dopamine;  GABA, gamma
aminobutyric acid; NAc, nucleus accumbens; NMDAR, N-methyl-
Recent advances in neuromodulation techniques and d-aspartate receptor; VTA, ventral tegmental area. Created with
devices led the indication of DBS to include movement Biorender.com
disorders, such as Parkinson’s disease, to neuropsychiat-
ric conditions, such as depression, Tourette’s syndrome,
and obsessive–compulsive disorder (OCD) [5–8]. The use and encoding a more biologically salient reward association
of DBS to reduce substance use has also been explored with surrounding environmental cues [13]. Such Pavlovian
in both preclinical and clinical settings [9–12], offering a drug-environmental conditioning can trigger the desire to
beacon of hope to patients suffering from medically refrac- seek more of the drug. For example, the drug associated cues
tory addictive disorders. This review aims to summarize can elicit a surge of dopamine, which may manifest in drug
and evaluate the potential therapeutic efficacy, mechanisms craving, seeking and ultimately use [14].
of action, and biomarkers involved in the use of DBS in Serotonin (5-HT) is another important neurotransmitter
the treatment of addictive disorders. We cautiously report mediating neuroplasticity, hedonic tone, motivational, and
that DBS is promising to treat certain refractory addictive reinforcement processes, as well as cognitive functions [15,
disorders. Recommendations for the future directions are 16]. 5-HT2A and 5-HT2C receptors in the prefrontal cortex
also provided. (PFC), NAc, and ventral tegmental area in particular appear
to be critical. 5-HT2A receptor increases, and 5-HT2C recep-
tor decreases, striatal dopamine release [15]. The seroton-
Addiction Neurobiology: a Brief Overview ergic neuroadaptations may be accountable for emotional
components such as anhedonia and depression during drug
Addiction is a highly complex process involving multiple withdrawal [16]. Recent evidence in mice also suggest that
neurochemicals and brain structures. A highly simplified serotonin signaling via 5-HT1B receptor may attenuate tran-
model highlights the central role of dopamine in mediat- sition from casual to compulsive cocaine use [17].
ing drug reward learning and drug seeking (compulsive) Chronic drug use ultimately alters the homeostatic neuro-
behaviors (Fig. 2). In essence, addictive drugs, such as the plasticity in the brain which not only results in a sensitized
psychostimulants and opioids, enhance synaptic concentra- dopaminergic system [18], but also leads to changes in the
tions of dopamine in forebrain subcortical structures, such glutamatergic system [13, 19, 20]. For example, calcium-
as the nucleus accumbens (NAc) [13]. These drugs of abuse permeable ionotropic glutamatergic AMPA receptors become
significantly enhance dopamine concentrations over and upregulated relative to glutamatergic NMDA receptors [13,
above the ability of natural rewards, hijacking the system 19, 20]. The changes in AMPA and NMDA receptors have

13
Deep Brain Stimulation for Addictive Disorders—Where Are We Now?

become a hallmark process of neuroplasticity that underpins Details of search criteria and inclusion and exclusion criteria
behavioral changes following chronic drug use [21]. Another are given in Supplementary Information Appendix A. Search
neurobiological adaptation that occurs with chronic drug use is results are presented in a Preferred Reporting Items for Sys-
the process of “dorsalization” [22]. Here, repeated exposure to tematic Reviews and Meta-Analyses (PRISMA) flow chart
drugs of abuse leads to recruitment of the circuit that projects (Fig. 3). This review was registered at the Open Science
from the orbitofrontal cortex (OFC) to the dorsal striatum (cau- Framework [38]. The process yielded 47 papers (25 pre-
date and putamen), where synaptic potentiation occurs at both clinical studies and 22 human studies), which were further
dopamine receptors 1 and 2 ­(D1-R and D ­ 2-R) on GABAergic reviewed in full text. The results of this literature search are
medium spiny neurons (MSNs). summarized in the sections below.
Activation of D­ 1-Rs receptors causes excitation in the post-
synaptic cell via cyclic adenosine monophosphate production
(cAMP) production [23] that ultimately leads to promotion Results and Discussion
of long-term potentiation [24]. ­D1-R- and NMDA recep-
tor–dependent activation of extracellular signal-regulated Preclinical Models and Clinical Correlation
kinase (ERK) in brain areas such as striatum is an important
pathway for drug-induced locomotor response and sensitiza- Animal DBS studies in the selected literature (Table 1)
tion [25–28]. For example, systemic injection of ­D1-R- or predominantly used conditioned place preference (CPP) or
NMDA antagonist prevents ERK phosphorylation triggered self-administration paradigms (Fig. 4) to model addiction.
by d-amphetamine in the mouse striatum [29]. In addition, Self-administration models have high construct, content, and
cocaine-induced ERK phosphorylation and locomotor sensi- face validity relative to human addictive behaviors. Exper-
tization is prevented with mitogen-activated protein kinases/ imenter-administration models such as CPP can measure
ERK kinase inhibitor or in mutant mice with alanine-replaced drug-induced behavioral sensitization, such as increased
Thr-34 residue of dopamine- and cAMP-regulated phospho- locomotion, and are easier to perform. Both models also
protein (DARPP-32) [29]. ­D1-R MSN structural plasticity allow modeling of withdrawal symptoms during abstinence
depends on NMDA and DARPP-32 signaling [24]. Psycho- and/or extinction phase, followed by relapse behaviors, such
stimulants appear to affect ERK signaling via DARPP-32- as cue or drug-induced reinstatement. Most common DBS
based ­D1-R- and NMDA intracellular pathways [29]. brain targets in the selected animal studies (Table 1) were the
Importantly, ­D1-R activation inhibits long-term depres- NAc [33, 39–45], PFC [44, 46, 47], and subthalamic nucleus
sion, as revealed by ­D1-R antagonist unmasking long-term (STN) [10, 37]. The following sections will focus on these
depression [30]. In contrast, D ­ 2-Rs can promote long-term three most studied targets.
depression and prevent the induction of long-term poten-
tiation [30]. However, ­D2-Rs are particularly sensitive to NAc Electrical Stimulation
dips in the tonic dopamine level, during which they activate
protein kinase A (PKA) to induce long-term potentiation The NAc is divided into core and shell subregions with dif-
[30–32]. Taken together, D ­ 1-R and D
­ 2-R are critical for bidi- ferent histopathological compositions, projections, and func-
rectional plasticity of striatal MSNs when dopamine levels tions [48]. Studies suggest that the shell is responsible for
are high vs low to guide adaptive and maladaptive behav- reinforcing properties of novelty, rewards (both drug- and
iors involved in reward learning [31, 32]. Therefore, ­D1-R non-drug-related), and drug relapse, whereas the core medi-
and ­D2-R signaling dynamically modulates cortico-striatal ates spatial learning, conditioned responses, responses to
plasticity depending on the level of synaptic dopamine to motivational stimuli, and impulsive choices [14, 49]. These
facilitate transition into persistent drug seeking and use. features of motivated behaviors suggest NAc shell under-
While DBS appears to target these processes involved in lying extinction and reinstatement behavior [50], and NAc
addiction to reduce drug seeking and taking [10, 33–37], core underlying the initial acquisition of drug taking and
the potential therapeutic efficacy and mechanisms of action cue-elicited drug seeking [48]. However, there is synergy
of DBS in the treatment of addictive disorders are poorly between the two subnuclei, and both play a role in drug
understood. We address this gap in the present review. addiction.
Stimulation of either the core or shell subregions of the
NAc using a range of low and high frequencies leads to
Methods reduced drug taking, seeking, or CPP in rats conditioned
with alcohol [34, 51, 52], nicotine [53], opioids [9, 37, 39,
A search was performed using keywords, such as “deep brain 41, 46, 54–57], cocaine [10, 33, 35, 40, 42, 43, 47, 58, 59],
stimulation” and “addiction” in MEDLINE, EMBASE, and or methamphetamine [45] (Table 1). Behavioral improve-
Cochrane Library database between 1974 and 6/18/2021. ment was related to the timing of when electrical stimulation

13
J. Yuen et al.

Fig. 3  Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) flow chart of literature search. Other sources include
those found by reviewing the references from included papers

was given—that is, stimulation given before a self-adminis- evidence in animal studies regarding the efficacy of NAc
tration session or CPP acquisition would reduce drug intake core vs shell stimulation. Both high- and low-frequency
or prevent CPP, respectively [39, 45]. When given during stimulation (HFS, LFS) of NAc (electrodes placed across
abstinence or reinstatement, stimulation would diminish core and shell) can reduce cocaine-primed drug seeking fol-
drug or cue-induced drug seeking after extinction and/or lowing 24 days of abstinence [43]. Other studies using core
forced abstinence phase [40–43, 45]. Such efficacy of stimu- as the DBS target showed reduced morphine-induced CPP
lation during abstinence to prevent relapse is particularly [39], ethanol self-administration [51], and heroin reinstate-
promising in the clinical context. ment [41]. The shell was also shown to be an effective tar-
While most animal and human studies (Table 2) show get for morphine-induced CPP [55], cocaine reinstatement
success in modulating addictive behavior using NAc stimu- [40], ethanol self-administration [44, 51, 52], and metham-
lation, there are two main issues. First, there is conflicting phetamine reinstatement [45]. In contrast, Vassoler et al.

13
Table 1  Summary of animal studies in deep brain stimulation and addiction, listed in chronological order. ACC​, anterior cingulate cortex; BLA, basolateral amygdala; CPP, conditioned place
preference; EA, experimenter-administration; EtOH, alcohol (ethanol); FR, fixed ratio; GABA, γ–aminobutyric acid; HFS, high-frequency stimulation; i.p., intraperitoneal injection; LFS, low-
frequency stimulation; LH, lateral hypothalamus; (m)PFC, (medial) prefrontal cortex; NAc, nucleus accumbens; OD, once daily; OFT, open field test; PAG, peri-aqueductal gray; pCREB, phos-
phorylated cyclic AMP-response element binding protein; SA, self-administration; s.c., subcutaneous injection; STN, subthalamic nucleus; vHipp, ventral hippocampus; VTA, ventral tegmental
area; NA, data not available

Author and year Species and strain Target; laterality Behavioral paradigm Stimulation parameters Main outcomes
(sample size)
Hz Pulse width Current DBS session

Pinel et al. Black-hooded male Amygdaloid Alcohol (1000– 60 NA 400 μA 45 stimulations Stimulation was performed to induce kindling
(1975) [85] rats (N = 27) complex; 2018 mg/kg ethanol) (1 s duration), (until displaying at least 1 seizure), and alcohol
unilateral (side via experimenter 3 × /day, 5 days/ was administered after stimulation. The
unspecified) intubation week withdrawal syndrome observed 9 h following
alcohol administration in kindled (stimulated)
animals was much more severe than that of the
controls
Beaulieu and Long-Evans male PAG; midline Subcutaneous morphine 50 NA 20–200 30 min after Large range of current applied to different rats
Thorn (1986) rats (N = 29) target 72 mg pellet (for 72 h) μA naloxone- (depending on analgesia threshold)
[54] vs placebo precipitated Stim. attenuated morphine withdrawal behaviors,
withdrawal esp. the recessive behaviors associated with
autonomic changes (e.g., eye-twitching) lasting
2 h or more
Deep Brain Stimulation for Addictive Disorders—Where Are We Now?

Levy et al. Sprague–Dawley Lateral Cocaine (1 mg/kg 20, 100 100 μs 200–400 30 min/day for DBS (100 Hz) in lateral hypothalamus or PFC
(2007) [35] male rats hypothalamus reducing to 0.5 mg/kg) μA 10 days reduced cocaine-, but not sucrose seeking. DBS
(N = 7–14/group) (100 Hz) or PFC self-administration. (100 Hz) in the PFC also reduced cocaine taking
(20 Hz, 100 Hz); Progressive ratio in a progressive ratio self-administration test
bilateral was also assessed at Cocaine-induced locomotor hyperactivity was
0.5 mg/kg exaggerated by lateral hypothalamus DBS but
Locomotion following was reduced by PFC DSB
cocaine challenge The behavioral findings were accompanied by
15 mg/kg (i.p.) GluR1 alterations in the NAc (increased) and the
VTA (variable depending on subregion)
Liu et al. (2008) Sprague–Dawley NAc core; right CPP induced by i.p. 130 210 μs 200–500 3 h (15-min trains Pre-conditioning DBS sessions prevented
[39] rats of unspecified morphine μA with 45-min morphine-induced CPP compared to sham
sex (N = 32) 10–60 mg/kg (increasing intervals)
dose)
Vassoler et al. Sprague–Dawley NAc shell or Cocaine self- 160 60 μs 150 μA Continuous DBS in NAc immediately following 10 or 20 mg/
(2008) [40] male rats dorsal striatum; administration during 2-h kg cocaine prime attenuated cocaine-induced
(N = 7–12/group) bilateral FR1 (250 μg/kg per reinstatement reinstatement compared to sham DBS. DBS in
infusion) progressing sessions dorsal striatum had no effects on reinstatement
to FR5
Knapp et al. Long-Evans male NAc shell or NAc Alcohol (0–10% ethanol) 160 200 μs 0–150 μA 5 min At 150 μA, but not at 100 μA or less, DBS in
(2009) [51] rats (N = 4–5/ core (2 groups); self-administration shell or core given prior to established alcohol
group) bilateral self-administration sessions reduced alcohol
consumption

13

Table 1  (continued)
Author and year Species and strain Target; laterality Behavioral paradigm Stimulation parameters Main outcomes
(sample size)

13
Hz Pulse width Current DBS session

Friedman et al. Sprague–Dawley Lateral habenula; Cocaine self- 10–100 500 μs 200 μA For 15 min at LFS alone appeared to have superior effect on
(2010) [58] male rats (N = NA) right administration start of task seeking behavior compared to HFS
0.25, 0.5, and 1 mg/kg With DBS consisting of a combination between 10
i.v. for 21 days and 100 Hz, drug seeking behavior was reduced
Reinstatement: 0, 5, 10, in self-administration, extinction and relapse
20 mg/kg i.p phases
Lesioning actually increases drug seeking behavior
(increased number of presses during extinction)
Tests controlled by forced swim test (depression-
like behavior), open-field test (locomotion), etc
Western blot showed elevated glutamatergic
receptor subunits NR1 and GluR1 and
scaffolding
Protein PSD95, but not GABA Ab, protein levels
in VTA with cocaine SA but normalized after
DBS
DBS reduced cocaine seeking behavior during
both self-administration and extinction training,
as well as lever-pressing behavior during
reinstatement
Henderson et al. Alcohol-preferring NAc shell; Home cage alcohol 140–150 60 μs 200 μA 1 h on 1 h off DBS given prior to established alcohol self-
(2010) [52] male rats (N = 20) bilateral (10% ethanol) self- DBS stage 2: administration sessions reduced alcohol
administration 24 h when free consumption and preference over water. DBS
access was given prior to alcohol-induced relapse following
allowed again 4–6 weeks of abstinence also reduced alcohol
consumption and preference over water
Rouaud et al. Long-Evans male STN; bilateral Cocaine self- 130 60 μs 50–130 DBS was applied DBS decreased cocaine but not food taking
(2010) [59] rats (N = 88) administration μA throughout Progressive ratio tests showed that DBS decreases
FR 1 (0.25 mg per each behavioral motivation to attain cocaine but increased
infusion) session motivation to attain food. Similar dissociation
Progressive ratio was was observed with CPP, with DBS reducing
also assessed at cocaine CPP but increasing food CPP
0.25 mg per infusion
CPP induced by cocaine
(10 mg/kg i.p.)
Guo et al. (2013) Sprague–Dawley NAc core; mixed Heroin self- 130 100 μs 0, 75, or 1-h continuous Daily DBS (75 and 150 μA) sessions during
[41] male rats (N = 54) unilateral or administration 150 μA stimulation abstinence reduced subsequent cue- and heroin-
bilateral FR1 (50 μg/kg per induced drug seeking, except for DBS only in the
infusion) paired with a left NAc. DBS increased pCREB but decreased
light cue ΔFosB in NAc core and shell
DBS did not affect locomotion, nor spatial learning
and memory
J. Yuen et al.
Table 1  (continued)
Author and year Species and strain Target; laterality Behavioral paradigm Stimulation parameters Main outcomes
(sample size)
Hz Pulse width Current DBS session

Ma et al. (2013) Sprague–Dawley NAc shell; Morphine EA 130 60 μs 2 V (stim. 60 min, 5 × /day CPP training was first given with increasing doses
[55] male rats (N = 25) bilateral 5 mg/kg i.p. voltages) for 30 days of morphine; naloxone then given to precipitate
progressing to “withdrawal.”
90 mg/kg i.p. over DBS prevented drug seeking behavior (in terms of CPP
12 days measurements) during re-instatement of morphine
Reinstatement— Place navigation studies were performed (Morris
2.5 mg/kg i.p water maze) to assess learning and memory—no
change by DBS
Pushparaj et al. Sprague–Dawley Insular cortex; Nicotine SA 130 90 μs 50 μA HFS started from DBS significantly attenuated nicotine taking, under
(2013) [53] male rats bilateral 0.03 mg/kg i.v. 5 min before both FR5 and progressive ratio schedules, as well
(N = 6–12/group) per infusion under a behavioral as nicotine seeking behavior induced by cues and
FR-5 then sessions and priming; no effect on food taking behavior
progressive ratio lasted throughout HFS of brain slices containing the insular region
schedule the sessions was found to inactivate insular neurons
Reinstatement dose
was 0.15 mg/kg s.c
Vassoler et al. Sprague–Dawley NAc shell or core; Cocaine self- 160 60 μs 150 μA Continuous DBS of NAc shell, but not core, immediately
Deep Brain Stimulation for Addictive Disorders—Where Are We Now?

(2013) [42] male rats bilateral administration during 2-h following 10 mg/kg cocaine prime attenuated
(N = 5–12/group) FR1 (250 μg/kg per reinstatement cocaine-induced reinstatement compared to sham
infusion) progressing sessions DBS (0 μA)
to FR5 DBS of NAc shell induced c-Fos expression
locally and in the infralimbic cortex
Wilden et al. Alcohol-preferring NAc shell; Alcohol (15% ethanol) 150 100 μs 100 or 200 65 min DBS (200 μA but not 100 μA) starting 5 min prior
(2014) [34] female rats (N = 7) unilateral self-administration μA to and throughout established daily alcohol self-
administration sessions reduced alcohol taking.
Alcohol taking resumed at baseline levels post
DBS sessions
Creed et al. C57BL/6 NAc shell; Locomotor sensitization 12 or 90 μs 50 μA 60 min DBS at 130 Hz, but not at 12 Hz, given
(2015) [33] transgenic male bilateral induced by cocaine 130 immediately before test transiently suppresses
mice (N = 6–12/ 20 mg/kg i.p locomotor sensitization. DBS had no behavioral
group) effects when applied 4 or 24 h before test
DBS at 130 Hz 24 h before did not affect cocaine-
evoked plasticity
DBS at 12 Hz in combination with a D ­ 1-R
antagonist reversed cocaine-induced
hyperlocomotion and plasticity. DBS at 12 Hz
had no effects by itself
Hamilton et al. Long-Evans male NAc; right Cocaine self- 20 or 100 μs 50–200 30 min A single DBS session did not affect cocaine taking.
(2015) [43] rats (N = 6–8/ hemisphere administration 160 μA Daily DBS 20 or 160 Hz sessions during 14 days
group) FR1 (500 μg/kg per of abstinence led to attenuated cocaine seeking
infusion) progressing following a 5 mg/kg cocaine prime when tested
to FR3 1 day, but not 14 days, after the last DBS session

13

Table 1  (continued)
Author and year Species and strain Target; laterality Behavioral paradigm Stimulation parameters Main outcomes
(sample size)

13
Hz Pulse width Current DBS session

Mehdipour et al. Wistar male rats Prelimbic cortex; Light–dark shock 60 NA 25, 20 min (duration Methodology was unclear (e.g., interval between
(2015) [46] (N = 6 in each right hemisphere passive avoidance 50, of 1 s every 5 s) doses and shock). Short-term stimulation was
group) reversed by morphine 100, or given
(10, 20, or 40 mg/ 150 μA DBS at 50 and 100 μA stimulation reversed
kg) i.p passive avoidance memory attenuation by
morphine. DBS at 25 and 150 μA had no effects
Hadar et al. Wistar male rats NAc shell, Home cage alcohol (5, 130 90 μs 5.25 V 24 h or 4 days Compared to sham DBS, 4 days of NAc shell
(2016) [44] (N = 6–15/group) infralimbic 10, or 20% ethanol) (stim. DBS led to augmented alcohol-induced relapse
cortex, or and water two voltages) following chronic abstinence. Acute (24 h)
striatum bottle choice self- DBS reduced alcohol-induced relapse. DBS in
(caudate, administration infralimbic cortex or striatum had no effects on
putamen); behavior. Acute NAc DBS (24 h) increased local
bilateral dopamine levels in alcohol-exposed rats
Martínez-Rivera Sprague–Dawley Ventral capsule/ Morphine EA 20 or 100 μs 100–200 60 min/day HFS of dorsal VS impaired extinction training
et al. (2016) male rats (N = 87) ventral striatum 5 mg/kg s.c./day for 130 μA over 9 days of and extinction memory but no effect for HFS of
[56] (VS); bilateral 4 days with CPP extinction ventral VS
training LFS of dorsal VS strengthened extinction memory
when tested 2 or 9 days after the cessation of
stimulation
Both HFS and LFS increased c-Fos expression in
infralimbic PFC, but only LFS increased c-Fos in
the basal amygdala and the medial portion of the
central amygdala
Batra et al. Wistar male rats NAc shell; Methamphetamine self- 130 60 μs 200 μA 3 h The first two daily sessions of DBS prior
(2017) [45] (N = 18) bilateral administration to self-administration sessions reduced
FR1 (0.05 mg/kg per methamphetamine taking compared to sham
infusion) DBS, while the remaining 3 DBS sessions had
no effects. These DBS sessions reduced cue-
induced reinstatement when tested 20–28 days
later
Wade et al. Wistar male rats STN; bilateral Heroin SA 130 60 μs 50–130 Throughout the STN HFS attenuated re-escalation of heroin intake
(2017) [37] (N = 32) 60 μg/kg i.v μA SA sessions post-abstinence in rats with extended access to
FR1 heroin
3- or 12-h sessions STN HFS inhibited substantia nigra,
5 days per week entopeduncular nucleus, and NAc shell
(measured with brain mapping analyses of
immediate-early gene expression) but not PFC,
and silenced STN neurons (measured using
recording ex vivo)
J. Yuen et al.
Table 1  (continued)
Author and year Species and strain Target; laterality Behavioral paradigm Stimulation parameters Main outcomes
(sample size)
Hz Pulse width Current DBS session

Chang et al. Sprague–Dawley Anterior insular CPP induced by 130 90 μs 150 μA 11 or 14 days 14 days of DBS during abstinence attenuated the
(2020) [57] male rats cortex; bilateral subcutaneous injection during expression of morphine-CPP but CPP relapsed
(N = 9–15/group) of morphine (10 mg/ extinction or 10 days after the cessation of DBS. Eleven
kg) abstinence days of DBS during CPP extinction facilitated
extinction acquisition as well as alleviating
morphine-induced reinstatement
OFT and novel object recognition test showed
no impairment to locomotion and recognition
memory
Proteomic analysis revealed the expression levels
of 8 morphine-regulated proteins in the anterior
insula were reversely changed by HF-DBS
Fakhrieh-Asl Wistar male rats Orbitofrontal CPP induced by 13 or 100 μs 150 μA 20 or 40 min, HFS but not LFS given daily for 3 days of
et al. (2020) (N = 108) cortex; bilateral subcutaneous 130 once daily conditioning significantly prevented CPP
[9] injections of morphine acquisition
3–7 mg/kg (increasing HFS but not LFS given daily for 6–10 days of
dose) extinction facilitated extinction acquisition and
Deep Brain Stimulation for Addictive Disorders—Where Are We Now?

blocked morphine-induced reinstatement


DBS did not cause any changes in locomotor
activity, novel objection recognition memory, nor
anxiety-like behavior
Guercio et al. Sprague–Dawley Infralimbic cortex, Cocaine self- 160 60 μs 150 μA Continuous DBS at BLA, vHipp, and infralimbic cortex
(2020) [47] male rats (N = 6–8/ prelimbic administration during 2 h immediately following 10 mg/kg cocaine prime
group) cortex, anterior FR1 (250 μg/kg per reinstatement attenuated cocaine-induced reinstatement
cingulate cortex, infusion) progressing sessions compared to sham DBS (0 μA), but BLA and
BLA or vHipp; to FR5 vHipp also attenuated sucrose reinstatement
bilateral Anterior cingulate cortex and prelimbic cortex
DBS did not attenuate either
Degoulet et al. Male Lister STN; bilateral Cocaine SA 8, 30, or 80 μs 50–150 DBS turned on 30 Hz but not 130 Hz DBS reduced pathological
(2021) [10] Hooded rats FR1 progressing to 130 μA and intensity cocaine seeking during treatment, whereas
(N = 84) random interval until was increased 130 Hz actually increased seeking behavior
120 s to reach acutely
Punishment sessions predetermined Compulsive, i.e., shock-resistant rats
(foot shock) also parameters exhibit pathological STN low-frequency
applied (rats divided prior to start oscillations (LFP) during cocaine escalation
into shock-resistant of behavioral (especially < 20 Hz)
and shock-sensitive session, and
subgroups) continued
during session

13
J. Yuen et al.

demonstrated that stimulation of the NAc shell but not the dopamine release in the NAc, which might explain the stim-
core attenuated cocaine-induced reinstatement [42]. ulation effect [69]. However, stimulation of the infralimbic
The contradicting evidence above may be explained by cortex starting 1 h or 4 days before alcohol prime did not
the large activation volume of stimulation pulses, such that affect alcohol-induced relapse following abstinence in rats
pulses centered on the core may also affect the shell, and [44]. The contrasting findings may be due to the different
vice versa. From human studies, the volume of activation mechanisms of actions of the drugs being given (alcohol is
was estimated to be between 30 and 120 ­mm3 [60] and the a depressant whereas cocaine is a stimulant). Alternatively,
human NAc volume is around 700 m ­ m3 [61]. However, infralimbic cortex stimulation may be more effective in
for adult Sprague–Dawley rats, the total volume of NAc is reducing relapse-like behaviors following extinction rather
approximately 5 m ­ m3 with the NAc core making up half than forced abstinence, consistent with the well-known role
of that [62]. Therefore, neighboring structures could be of the infralimbic cortex in extinction [70]. Infralimbic cor-
co-stimulated in rats. Notably, the core and shell divide is tical DBS success is concordant with reduced craving by
functionally present in humans while they are less distinct transcranial direct current stimulation (tDCS) of dorsolat-
in rodents [63, 64]. eral PFC in humans [71]. However, DBS of the prelimbic
The second issue concerns laterality. While most studies cortex following cocaine priming had no effect on cocaine
used bilateral stimulation, some studies only used unilateral reinstatement in rats [47].
NAc stimulation [34, 39, 41, 43]. One study used left, right, In the OFC, DBS prevented rats from learning morphine-
and bilateral NAc core stimulation, demonstrating only the induced CPP, while DBS during extinction facilitated extinc-
latter two were effective in reducing heroin seeking behavior tion acquisition and prevented morphine-induced reinstate-
[41]. It is unclear why such asymmetry exists. Dopamine ment [9]. Chronic DBS of the anterior insular cortex either
concentration in the NAc is reported to be higher on the side during 11 days of extinction or 14 days of abstinence reduced
ipsilateral to the paw-dominance of a mouse [65]; however, CPP, but CPP relapsed 10 days after chronic DBS [57]. This
morphometric NAc human studies based on laterality have finding is consistent with how repetitive transcranial mag-
been controversial [49]. netic stimulation (rTMS) reduced heavy drinking only dur-
Almost all of these published studies in humans show ing the intervention but not after in alcohol-dependents [72].
some improvement from DBS in the NAc in addiction- The insula’s superficial location and delicate anatomy may
related behaviors (Table 2) as well as cognitive improvement, be better suited for non-invasive interventions such as rTMS
which raises the possibility of potential publication bias. rather than DBS.
With this precaution in mind, NAc DBS is likely the low- Anterior cingulate cortex (ACC) dysfunction can disrupt
est hanging fruit for refractory addictive disorders. A recent ongoing processing and detection of erroneous outcomes
systematic review supports NAc DBS to treat substance relevant for reward learning [73, 74]. ACC DBS following
use disorders, demonstrating that NAc DBS in patients led cocaine prime did not alter reinstatement following self-
to a relapse rate of ~ 39% with a reasonable safety profile, administration in rats [47]. However, when people with
which is an improvement compared to the ~ 60% relapse rate alcohol dependence received bilateral DBS (6- or 10-Hz
reported in that population (selection bias factors notwith- stimulation), craving was significantly reduced [75]. The
standing) [66]. In addition, individual case reports revealed reduced craving was associated with a reduction in beta-1
that those with comorbid psychiatric disorders, such as band (13–18 Hz) current density on electroencephalogram
depression and OCD, showed improvement in these other (EEG) post-stimulation in the rostrodorsal ACC [75]. Alco-
symptoms with NAc DBS with or without anterior limb of hol craving also correlated with beta activity in the dorsal
internal capsule (ALIC) intervention (DBS or capsulotomy) ACC in a case report, although relapse was associated with
[12, 67, 68]. DBS may provide additional benefit for patients increased gamma band activity [76]. Another case report
with multiple psychiatric comorbidities. showed that a patient with concomitant OCD and alcohol
dependence was successfully treated with ACC rTMS [77].
Interestingly, NAc DBS reduced craving correlated with
Cortical Stimulation improvement in error-related negativity in LFP in the ACC
in humans [78], suggesting the effect of NAc DBS may alle-
The PFC is believed to provide top-down control of NAc viate ACC dysfunction. In addition to DBS, rTMS of the
in addiction (Fig. 2). DBS of the PFC (subregion not speci- dorsal ACC demonstrated improvement in alcohol craving
fied) significantly reduced cocaine seeking and motivation and concomitantly reduced NAc activation on fMRI [76].
to take cocaine measured by progressive ratio test [35]. Taken together, DBS in ACC may be particularly relevant
Stimulation of the infralimbic cortex of the PFC following for alcohol use.
a cocaine prime also reduced cocaine reinstatement [47]. Most of these cortical regions project to the NAc, rais-
Notably, mPFC stimulation in rats showed increased phasic ing the possibility that their DBS effects are mediated

13
Deep Brain Stimulation for Addictive Disorders—Where Are We Now?

via these connections [49, 79]. Animal studies show not


only strong projections from the cortex to NAc, but also a
high degree of interplay between the two regions [80]. For
example, c-Fos reactivity is observed in infralimbic cortex
and NAc during DBS [42, 56] Pharmacological silencing
of cortical interneurons can reinstate drug seeking [42, 81],
which is consistent with silencing effects observed in the
NAc [81]. Nevertheless, NAc is unlikely to be the sole
driver of effects of DBS in the PFC considering that DBS
in the prelimbic cortex does not affect drug seeking in rats,
even though prelimbic cortex has strong connections to
NAc [47].
Fig. 4  Illustration of two most widely used experimental para-
digms for the study of drug addiction in animals. On the left, con-
STN Stimulation ditioned place preference (CPP) is shown, where “non-contingent”
drug-administration is associated with one side of the chamber dur-
Parkinson’s patients who take dopaminergic medications for ing drug-side associated conditioning. If the drug is experienced as
rewarding, the animal will spend more time in the drug-administered
their movement disorder may chronically feel compulsion to side relative to the other side, even in the absence of further drug
over-consume such agents, even at the expense of dyskine- administration. On the right, “contingent” drug self-administration
sia and other side effects. Studies suggest that STN DBS in paradigm allows the animal to regulate its own drug intake (e.g., via
humans may improve movement symptoms as well as reduce nose-poke or lever-pressing). A discrete light and/or tone cue may be
provided in association with the drug intake to serve as a drug-associ-
compulsive use of medications [82, 83]. ated cue. In extinction, the animal is placed back into the environment
STN DBS or lesions in rats have been shown to reduce where CPP or self-administration is acquired but without drug avail-
motivation for cocaine taking, while increasing motivation ability. Extinction sessions lead to reduced place preference or drug
to consume more common rewards such as food [10, 37, 59, seeking. During reinstatement, the drug, the drug-associated cue,
and/or stress can be given, which can lead to “relapse” measured by
84]. The lesion study in particular suggests that STN DBS preference or drug seeking. Created with Biorender.com
may work via local inactivation [59, 84]. Further evidence
shows STN HFS in rats inhibiting substantia nigra, entope-
duncular nucleus, and NAc shell measured with brain map- High‑ Versus Low‑Frequency Stimulations
ping analyses of immediate-early gene expression but not
PFC, and silencing STN neurons measured using recording Whereas most animal studies reported HFS (> 130  Hz)
ex vivo [37]. effects on behavior, some used a lower frequency (< 20 Hz)
Although HFS of STN in rats reduced both cocaine tak- and observed beneficial effects targeting the NAc [43], lat-
ing and heroin seeking [37, 59], a conflicting study demon- eral habernula [58], ventral striatum [56], and periaque-
strated only LFS (30 Hz) but not HFS modulates cocaine ductal gray [54] even at frequencies as low as 8 Hz (STN)
taking behavior in the presence of foot-shock punishment [10] (Table 1). In contrast, LFS targeting the NAc shell [33],
[10]. Abnormal alpha/theta and low-beta oscillatory activ- PFC [35], or OFC [9] were ineffective. Interestingly, DBS
ity during escalation of the cocaine intake phase also pre- at 12 Hz alone did not affect cocaine-induced locomotor
dicted the subsequent emergence of compulsive-like seeking sensitization, but combined with dopamine D ­ 1-R antagonist,
behavior, where the animals were shock-resistant [10]. The reversed cocaine-induced hyperlocomotion and plasticity in
discrepancy here may be explained by the absence/presence the NAc shell [33]. Such findings suggest that LFS may be
of punishment during drug taking, but further studies are effective only when basal dopamine signaling is low.
warranted to understand STN LFS vs HFS outcomes. The effect of HFS in NAc shell may be short-lived, lasting
less than 4 h in a cocaine experimenter-administered mouse
Other Brain Regions model [33] or 1 day in an alcohol self-administration rat model
[34]. However, reduction in cue-induced seeking behavior was
Electrical stimulation targeting other brain regions in ani- maintained more than 4 weeks post-HFS (NAc shell) com-
mals—amygdala [47, 85], hippocampus [47], periaqueductal pared to the sham group in rats that self-administered meth-
gray matter [54], lateral hypothalamus [35], dorsal striatum amphetamine [45]. Yet, LFS and HFS (mixture of NAc core
[40, 44], and lateral habenula [58]—has also been described and shell) effects lasted at least 2 weeks but less than 30 days
in the literature (Table 1). However, due to the limited num- in a cocaine self-administration rat model [43]. The different
ber of studies per brain region, meaningful conclusions are DBS parameters, targets, drugs, rodents, and tests across these
difficult to draw. Hence, these regions are beyond the scope studies (Table 1) pose a challenge to evaluate the best DBS
of this review. protocol to reduce drug use.

13

Table 2  Summary of human studies, listed in chronological order. ACC​, anterior cingulate cortex; ALIC, anterior limb of internal capsule; AMP, amphetamine; BNST, bed nucleus of stria ter-
minalis; BZ, benzodiazepine; CT, computed tomography; EEG, electroencephalogram; EtOH, alcohol; F, female; LFP, local field potential; M, male; MRI, magnetic resonance imaging; NAc,
nucleus accumbens; OCD, obsessive–compulsive disorder; PD, Parkinson’s disease; PET, positron-emission tomography; QoL, quality of life; STN, subthalamic nucleus; VC/VS, ventral cap-
sule/ventral striatum; NA, data not available

13
Author and Indication for Target Drug Stimulation parameters Laterality Adverse events Outcome
year DBS (N; age
sex) Hz μs V

Witjas et al. PD (2; 53 M, STN Dopaminergic High NA NA Bilateral None reported Drugs stopped or much reduced immediately post-
(2005) [82] 38 M) agent (despite operation
severe At 2 years’ follow-up, continue to be show benefit
dyskinesia) Dyskinesia and mood also much improved with no
change in cognition
Kuhn et al. Addiction (1; NAc EtOH 130 90 4.5 V Bilateral None reported Comorbid anxiety and depressive disorders
(2007) [141] 54 M) Reported feeling of “inner appeasement” acutely
After 12 months of treatment, the alcohol consumption
and craving reduced dramatically
Kuhn et al. Tourette’s NAc Nicotine 130–145 90, 180 3–6 V 5 bilateral, 5 None reported Fagerström Test for Nicotine Dependence was used to
(2009) [93] syndrome unilateral assess smoking behavior
(4), OCD (5) At 30-month follow-up, 3 out of 10 stopped smoking
or anxiety but the others mostly had no changes
disorders Those who quit smoking had higher stimulating voltages
(1) (10; but difficult to compare due to small numbers
7 M and 3F,
range from
28–58 years)
Mantione et al. OCD (1; 47F) NAc Nicotine 185 90 3.5 V Bilateral None reported Comorbid mild anxiety and depression
(2010) [94] Dramatic improvement in OCD, anxiety, and depression
Smoking cessation and weight loss was observed after
10 months post-operation (up to at least 2-year follow-up)
Kuhn et al. Addiction (1; NAc EtOH 130 120 5.5 V Bilateral None reported Abstinence after 1 year of treatment
(2011) [78] 69 M) Improvement seen in psychometric score
EEG showed enlargement in error-related negativity
(amplitude when error occurs during test) and
reduction in error rate
Zhou et al. Addiction (1; NAc Heroin 145 90 2.5 V Bilateral Transient Abstinent for 6 years from implantation (removed after
(2011) [90] 24 M) confusion first 3 years)
and urinary Improvement in cognition and memory
incontinence
initially
Heldmann Addiction (1; NAc EtOH 130 90 3.5 V Bilateral None reported Risk taking behavior was assessed by gambling task—
et al. (2012) 38 M) less risk taking behavior when DBS was switched on,
[92] compared to off-state
PET showed activations in the paracingulate cortex, temporal
poles, precuneus and hippocampus with active DBS
Except for the temporal pole, these activations were not
seen when DBS was off
J. Yuen et al.
Table 2  (continued)
Author and Indication for Target Drug Stimulation parameters Laterality Adverse events Outcome
year DBS (N; age
sex) Hz μs V

Valencia- Addiction (1; NAc Heroin 180 90 3.5 V Bilateral None reported Mostly clean for 6 months apart from self-limiting
Alfonso 47 M) relapse (although activation of some contacts initially
et al. (2012) increased craving)
[142] Intracranial EEG showed higher power for drug-related
compared with non-drug-related cues in the gamma-
band (40–60 Hz), which also has a lower correlation to
frontal theta signal compared with the drug-unrelated
responses on the right side
Voges et al. Addiction (5; NAc EtOH 130 90 3.5 V Bilateral Transient Average follow-up was 38 months
(2013) all male, hypomania LFP using externalized electrodes showed error-related
[143] Cases range from modulations at NAc during neuropsychological
overlap with 36–65 years) task and error-related negativity obtained from NAc
Muller et al. preceded the surface error-related activity
(2009) [144] All subjects experienced significant improvement of
and Heinze craving. Two remained completely abstinent for more
et al. (2009) than 4 years
Deep Brain Stimulation for Addictive Disorders—Where Are We Now?

[145]
Kuhn et al. Addiction (2; NAc EtOH and AMP, 140 120 5 V Bilateral 1 seizure (known Improved depressive and anxious symptoms, as well as
(2014) [67] 33 M, 31F) AMP and BZ 130 90 4.5 V epilepsy) consumption and craving in methadone and heroin
Patients started to consume other psychotropic
substance and continued to consume amphetamine out
of “boredom” and for weight-related reason without
subjective craving
Gonçalves- Addiction (1; NAc, BNST, Cocaine 130 150 2.5–4 V Bilateral Only transient, Previous comorbid heroin addiction
Ferreira 36 M) ALIC e.g., metallic DBS at ALIC showed no efficacy but posterior NAc and
et al. (2016) taste, warm BNST appeared to be effective targets
[146] sensation at high After 2.5 years, including a blinded on/off period, DBS
voltages resulted in improvement in cocaine dependence both
objectively and subjectively
Müller et al. Addiction (5; NAc EtOH 130 90 3.5–4.5 V Bilateral None reported All patients reported complete absence of craving for
(2016) [11] all males, Initial alcohol
Cases (3) range transient Follow-up was up to 8 years (2 died after 4 and 8 years)
overlap with from 35 to hypomania Two remained abstinent for years and 3 showed a
Muller et al. 55 years) reduction of consumption
(2009) [144]

13

Table 2  (continued)
Author and Indication for Target Drug Stimulation parameters Laterality Adverse events Outcome
year DBS (N; age

13
sex) Hz μs V

Ge et al. Addiction (7; NAc +  Heroin 145 150–240 2.0– Bilateral None reported Abstinence ranges from 3 months (lost to follow-up)
(2018) 6 × M and ALIC (NAc) 3.3 V to at least 40 months (4 out of 7 have been abstinent
[107]—5 1 × F, range 185 for > 20 months); mood also improved for most
patients from 26 to (ALIC) patients
overlap with 50 years) Pre-DBS LFP: Both NAc and ALIC showed prominent
Chen et al. theta and alpha band activity
(2019) [12] A distinct beta band peak was also detected at ALIC
There was a significant negative correlation between
the power of theta band of ALIC and craving score,
and a positive correlation between the power of alpha
band of NAc and scores on the Hamilton depression
inventory
LFPs of the NAc and ALIC exhibited higher coherence
in the theta and alpha bands
Zhang et al. Addiction (1; VC/VS Heroin 130 90 3.5 V Bilateral Hypomania Comorbid antisocial personality disorder (baseline)
(2018) [132] 39 M) Death secondary Initial improvement but multiple relapses despite
to overdose altering DBS parameters
(after relapse)
Chen et al. Addiction (8; NAc +  Heroin 130–185 150–240 1.5–7 V Bilateral One intracranial Some had comorbid depression and OCD
(2019) 7 × M and ALIC hemorrhage Five patients were abstinent for more than 3 years, and
[12]—5 1 × F, range (< 3 mL) with the other 2 relapsed after abstaining for 6 months
patients from 22 to no neurological 1 was lost of follow-up at 3 months
overlap with 50 years) deficit Craving reduced if the patients remained abstinent.
Ge et al. Transient fever, Simultaneous DBS of NAc and ALIC also improved
(2018) [107] headache, the quality of life, alleviated psychiatric symptoms,
insomnia, and and increased glucose metabolism in certain
subjective slight addiction-related brain region (e.g., inferior frontal
memory decline gyrus) on PET scan
Ge et al. Addiction (2; NAc Methamphetamine 150, 165 210, 240 2.5, 3.3 V Bilateral Insomnia, Only one out of two patients remained abstinent (at 1.5
(2019) [147] 38 M, 49 M) hypomania, to 2.5 years), and reviewing post-operative CT and
teeth grinding at pre-operative MRI showed incorrect placement of lead
higher voltages in the subject who relapsed
Leong et al. Addiction ACC EtOH (refractory 6 or 10 NA NA Bilateral Infection (2), The self-reported alcohol craving reduced by 61% (end
(2020) [75] (8; 4 × M (rostrodorsal) to multiple Mode: Burst seizure (1), point ranged from 12 to 48 weeks, depending on
Cortical and 4 × F, treatments) (7) or tonic venous infarct presence of adverse events)
implant 20–65 years) (1) (1), psychosis Post-stimulation led to reduction in in current density at
(1), impulsive the rdACC for beta-1 band (13–18 Hz) on EEG
behavior (1)
J. Yuen et al.
Table 2  (continued)
Author and Indication for Target Drug Stimulation parameters Laterality Adverse events Outcome
year DBS (N; age
sex) Hz μs V

Sildatke et al. Addiction (4; NAc Opioid NA NA NA Bilateral It appeared DBS EEG and LFP studies showed significant accuracy-
(2020) [148] all males, was not turned related modulations in the LFPs at the time of the
range on but the error-/correct-related negativity in 2 patients and at the
from 24 to electrodes were time of error-positivity in 3 patients (Eriksen flanker
57 years) used for LFP task)
recording
Zhang et al. Addiction (1; NAc Heroin NA NA NA Bilateral None mentioned Reduction in drug cravings and remained drug-free
(2020) [91] 42 M) (except for 1 episode at 6 months) at 1 year
Implants were removed at the end of treatment (at
patient’s request)
Zhu et al. Addiction (1; NAc DBS Bucinnazine, 160 90 2.70, Bilateral None reported Comorbid depression and anxiety disorder
(2020) [68] 28 M) combined morphine, 2.75 V Reduced craving since 3 months and abstinent for at
with anterior hypnotics least 1 year
capsulotomy (13 years) Improvement in sleep, cognition, depression/anxiety,
QoL seen
Mahoney et al. Addiction (1; NAc/ALIC Opioid and BZ 145 90 6 V Bilateral None Abstinent at 12 months
Deep Brain Stimulation for Addictive Disorders—Where Are We Now?

(2021) [149] 30sM) Improved subjective craving, mood, frontal, and


executive function
PET scan showed an increase in glucose metabolism
in the dorsolateral prefrontal and medial premotor
cortices

13
J. Yuen et al.

Potential Mechanism(s) of Action the NAc shell led to antidromic stimulation of the cortex via
cortico-accumbal afferents [42]. Consistent with infralimbic
Neuronal Inhibition cortex-NAc circuitry in DBS, electrophysiological studies
showed NAc HFS but not LFS enhances both NAc-evoked
Despite the promising results of DBS in addiction, the LFP responses and spontaneous LFP slow oscillatory activ-
mechanism of action is poorly understood and likely highly ity in OFC in an antidromic fashion [87]. These results fur-
complicated. Several hypotheses on the mechanism of DBS ther demonstrate the complex interplay between NAc and
in addiction have been suggested, such as direct inhibition/ the cortex.
excitation of neural activity, information lesioning, and
synaptic filtering where DBS blocks transmission of patho- Dopamine Replacement
logical low-frequency oscillations [86]. In particular, the
inhibition hypothesis is supported by animal studies where One study showed that HFS of the NAc shell led to a para-
both injection of GABA agonists (i.e., temporary inhibition) doxical augmentation in relapse behavior in a rat model of
and HFS in the NAc was found to reduce alcohol use in alcohol addiction [44], a contrary finding to other studies
rats [34]. Similar effects were also seen between STN lesion (Table 1). The authors also demonstrated an increase in
paradigms and DBS with cocaine conditioning [59, 84]. It extracellular levels of dopamine as measured by microdialy-
is likely the neurons and their respective synaptic activities sis. DBS-evoked increase in dopamine is reminiscent of the
respond in a combination of different ways depending on the intracranial self-stimulation model, where the subjects self-
stimulation frequency and the drug being tested. administer electrical stimulation via implanted electrodes in
the brain [88]. Reinforcing effects of electrical stimulation
Depotentiation of Excitatory Inputs onto ­D1‑R Via is presumably mediated by dopamine release in the reward
Modulation of Glutamate Receptors circuitry [89]. In addition, medial PFC DBS evoked dopa-
mine release in the NAc in a frequency-dependent manner
Evidence also suggests the interaction between dopamine in anesthetized rats measured by fast-scan cyclic voltamme-
and glutamate receptors is involved in the mechanism of try (FSCV) [69]. Taken together, DBS of PFC may operate
DBS action. For example, NAc shell LFS (12 Hz), only partially via the replacement of dopamine, but whether this
when used in combination with a D ­ 1-R antagonist, could can reduce drug taking or seeking has yet been tested. Most
reverse the motor sensitization and cocaine-evoked plastic- of the DBS studies did not employ in vivo microdialysis
ity changes in ­D1-Rs on GABAergic MSNs, in the form of techniques for direct quantification; hence, replacement of
increases AMPA/NMDA glutamate receptor ratio [33]. By dopamine as a DBS mechanism is unconfirmed.
using a metabotropic glutamate receptor (mGluR1) antago-
nist to block the therapeutic effect, the authors proposed that Limitations of Animal Models
the mechanism of action of DBS is mGluR-dependent, the
activation of which depotentiates excitatory synaptic inputs Human subjects tend to require weeks and months of DBS
onto ­D1-Rs on GABAergic MSNs [33]. Such depotentiation to observe an improvement, whereas animal studies show an
was associated with a normalization in drug-adaptive behav- almost immediate effect. In animal studies, the condition-
ior. The findings are also consistent with the upregulation of ing occurs over days (rather than years). The short time-
GluR1 seen in the NAc in other stimulation studies [35, 36]. frame indicates that the neuroplasticity reversal may not be
In contrast, both LFS (20 Hz) and HFS (160 Hz) stimula- the dominating effect of DBS in animals and the dopamine
tion of the NAc core attenuated cocaine seeking behavior in replacement caused by stimulation may mediate the acute
rats without the need of additional medications [43], similar improvement in behavior. In humans, DBS likely operates
to the outcome of PFC stimulation [35]. The contrasting via reversal of neuroadaptations that have been built up on a
findings between NAc core and PFC stimulations may be much longer time scale than animals subjected to condition-
explained by the different levels of basal dopamine in these ing. This interpretation is supported by case reports where
brain regions. Examining how DBS affects mGluR1 expres- subsequent explantation of the DBS device did not reverse
sion in different regions to affect D­ 1-R signaling would be the initial effect of abstinence [90, 91]. In addition, a fur-
informative in future work. ther case report showed that NAc DBS reduced risk taking
behavior and increased activation of brain regions implicated
Antidromic Activation of Cortex in behavioral control arising from lasting neuroadaptations,
measured by positron-emission tomography [92].
Post-mortem brain c-Fos immunohistochemistry following Some human studies also include subjects with comorbid
in vivo NAc shell HFS showed both local activation and OCD, depression, and anxiety [12, 93, 94]. In fact, addiction
activation of the infralimbic cortex, suggesting that DBS of may be part of a maladaptive coping mechanism for other

13
Deep Brain Stimulation for Addictive Disorders—Where Are We Now?

underlying disease/s. DBS may alleviate addiction by reduc- tailored to maximize therapeutic efficacy while minimizing
ing the severity of an underlying comorbid disease, which side effects. Nonetheless, a recent systematic review showed
is challenging to model in animals. For example, the higher that NAc DBS has similar efficacy and safety profile as radi-
cognitive and emotional component as well as the impact of ofrequency ablation of the NAc [66].
social cues are difficult to assess in animals.

Alternatives to DBS Future Directions

Given the invasive nature of DBS, other neuromodulatory Potential Biomarkers


modalities are worth considering. Of note, noninvasive
techniques such as rTMS and tDCS have been of particu- The hunt for a reliable biomarker serves three critical pur-
lar interest [95, 96]. A detailed review of these techniques poses: to facilitate patient selection, to monitor the progress
and outcomes is out of the scope of this article but in brief, of the disease (and therapeutic effect of treatments), and
rTMS operates by applying a stimulating coil at the scalp, to progress towards a holy grail of DBS—a closed-loop
which generates magnetic fields. The magnetic fields then therapeutic system. Not only can this potentially increase
pass through the skull and induce strong focal currents in the the battery longevity (hence reducing the need for battery
underlying brain tissues [97]. It is believed that long-term replacement), it also can increase the efficacy of stimula-
repetitive stimulation can lead to neuroplasticity, which can tion by tailoring to the patients’ neural responses. Further, a
provide therapeutic effect to the addicted brain [97]. tDCS “tolerance” effect can be observed with chronic open-loop
involves application of a direct or alternative electrical cur- DBS stimulation in movement disorder patients, with the
rent to the scalp, such that current travels from a positive stimulation effect wearing off after a period of time [105].
(cathode) to a negative (anode) electrode. Cortical neurons Such a tolerance is also likely in patients receiving DBS to
are suggested to be facilitated under the anode, whereas treat psychiatric illnesses. Closed-loop system with inter-
those under the cathode are suppressed [98]. Although rTMS mittent, dynamic stimulation can potentially minimize tol-
and tDCS have been shown to be effective in preclinical erance. Such biomarkers need to be easy to measure and
studies and small clinical case series on reducing symptoms possess a relatively high spatiotemporal resolution of meas-
such as craving, more data are required to ascertain their urement that, in turn, correlates well with clinical/behavio-
long-term safety profiles, efficacy, and the ideal patient pro- ral changes. In our opinion, the most promising candidates
file [98–100]. Although research shows that non-invasive would be electrophysiological and neurochemical measure-
neuromodulation can be successful in certain cases with- ments, although they will need to be tailored towards indi-
out the need for neurosurgery, the main limitations are the vidual addictive substances and patients.
focality of these non-invasive treatments and the relatively In terms of electrophysiological measurements, LFP is
low spatial resolution. These non-invasive interventions also the most widely studied potential biomarker among DBS
require numerous frequent visits to facilities in a specialized, studies. LFP’s wide use is mostly because of its measure-
tertiary center. In contrast, DBS can be implanted as a per- ment using the stimulating electrodes themselves. In ure-
manent or chronic stimulation system, which can potentially thane-anesthetized rats, NAc HFS suppressed pyramidal cell
be programmed remotely [101]. firing and enhanced slow LFP oscillations in the OFC via
There was a recent success of semi-invasive MRI-guided antidromic activation of cortico-striatal recurrent inhibition
focused ultrasound (MRgFUS) thalamotomy in the treat- [87]. In addition, prolonged NAc HFS and LFS (delivered
ment of movement disorders and other psychiatric disorders over 90 min) have been shown to affect both spontaneous
(depression and OCD) [102, 103]. Such a technique would and evoked LFPs in the medial PFC, lateral OFC, mediodor-
avoid the complications associated with the implantation of sal thalamus (MD), and NAc sites [106]. HFS also produced
devices, which may necessitate explantation for reasons such widespread increases in spontaneous beta and gamma power
as lack of efficacy or infection. Alongside DBS, MRgFUS and enhanced coherent beta activity between MD and all
offers a novel therapeutic avenue that should be explored to other regions. In contrast, LFS elevated theta power in the
treat refractory addictive disorders. MD and NAc. Acutely, HFS increased and LFS decreased
Invasive techniques can offer a more focal region of ther- induced relative gamma coherence. Stimulation-induced
apy. Ablation techniques in drug addiction targeting the NAc changes in gamma coherence shows that NAc DBS may
with promising results have long received attention [104]. achieve therapeutic effects by restoring the synchronicity
Indeed, a recent systematic review showed that NAc DBS of a neural circuitry that is disrupted in neuropsychiatric
has similar efficacy and safety profile as radiofrequency diseases. The differences in the effect on LFPs between
ablation of the NAc [66]. Compared to ablation techniques, HFS and LFS may also explain the differences in behavioral
however, DBS allows adjustment of stimulation parameters effects described above.

13
J. Yuen et al.

In a study assessing 7 people with heroin use disorder, and 2), and the use of larger animals—preclinical studies
pre-DBS electrophysiology revealed prominent theta and are predominantly rodent-based, but also well suited to large
alpha band activity in both the NAc and ALIC [107]. Addi- animal models. In addition, we have focused on technologies
tionally, a beta band peak was detected in the power spec- that are currently clinically viable in this review, namely
tra of ALIC LFPs, which, the authors hypothesized, may electrical DBS devices. However, the use of optogenetics,
represent the activity of striatal bridge cells. Furthermore, where a viral vector is able to transfect a specific neural
there was a negative correlation between the power of the population, is increasingly popular in the preclinical study
theta band of ALIC LFPs and subjective craving, and a of underlying pathophysiology of different neuropsychiatric
positive correlation between the power of the alpha band of disorders and relevant therapies. Optogenetics has provided
NAc LFPs and depressive symptoms. LFPs of the NAc and new insights into DBS protocol optimization for Parkinson’s
ALIC both exhibited higher coherence values in the theta disease with translational potential [123, 124], which may
and alpha frequency bands. No comparison with post-DBS be extended to psychiatric illnesses. In addition, recent opti-
recordings was given in this study. Together with the animal cal imaging techniques allow combination of fiber photom-
and human DBS studies on NAc [108, 109], the most con- etry and electrophysiology to study specific cell population
sistent biomarker frequency is yet unknown. Further studies activity via genetically encoded calcium indicators, includ-
are required for clarification. New technologies such as the ing during deep brain stimulation [125, 126]. Cell-specific
Medtronic Percept Stimulator [110] may be able to provide viral transfection in calcium imaging or optogenetics helps
more information by simultaneously recording from and to study specific ion channels or neurotransmitters. How-
stimulating brain targets in humans. In addition, trials inves- ever, the use of viruses in humans is very limited. Further
tigating electrophysiological changes secondary to DBS in technological advancements are necessary to harness optical
addiction (such as [111]) will provide more insights. methods in the clinical setting.
Unlike electrophysiological signals (i.e., action poten- Currently, the use of DBS in addiction remains off-label,
tials) that naturally lead to neurotransmitter release into primarily due to the lack of high-quality clinical evidence.
synapses, quantification of synaptic-related neurotransmit- As seen in Table 2, most human studies consist of observa-
ter extracellular concentrations directly measures the activa- tional studies with a small sample size. However, more clini-
tion of relevant neural circuits (such as the ones depicted in cal trials are currently being conducted. The ClinicalTrials.
Fig. 2). Most studies in addiction focus on the monoamines, gov database show that there are currently around 10 DBS
particularly dopamine, glutamate, and 5-HT. One animal trials underway trying to tackle this paucity of scientific evi-
study reviewed here [44] employed in vivo microdialysis dence, targeting the NAc, ALIC, and STN [127]. As dem-
and found that local dopamine concentrations increased after onstrated in the present review, they are all very promising
NAc shell HFS. While in vivo microdialysis has long been targets. In addition, we would add PFC to this list given the
the gold standard for sampling neurochemicals, the process success from animal studies [47] as well as human imaging
necessitates an external analyzer and has low temporal reso- studies showing its dysfunction in subjects with substance
lution. In contrast, in vivo electrochemical techniques, such use disorder [128].
as FSCV, provide potential benefits such as higher temporal Supporting the potential of DBS in alleviating addic-
resolution, less tissue damage, and the ability to be chroni- tion-related behaviors, DBS to treat obesity has also shown
cally implanted [112–116]. However, compared to micro- early successes [129–131]. Given the similarities between
dialysis, electrochemical techniques have other limitations. substance use disorders and obesity, the findings in DBS
Often, they are restricted to measurement of a single electro- for substance use disorder presented so far may be translat-
active amine at a time, such as dopamine or serotonin. Thus, able to DBS for obesity. With the increasing prevalence and
future studies which aim to develop biomarkers for DBS comorbidities associated with excessive calorie consumption
efficacy will need to demonstrate suitability of neurochemi- in the modern society, this can be hugely beneficial.
cal biomarkers for translation and may be limited by present While it is exciting to perform cutting-edge patient sur-
detection technology. While microdialysis provides impor- geries, it is also important to recognize that DBS is not
tant measurements of the tonic (basal) extracellular levels an immediate cure and requires careful patient selection,
of neurochemicals, voltammetry can provide measurement consent, and patient-based optimization (including treating
of phasic and tonic measures of neurochemicals [117–122]. other comorbidities); otherwise, DBS can lead to a risk of
major adverse outcomes [132]. DBS also has cost, transla-
Future Preclinical and Clinical Research Considerations tional, and capacity limitations for a highly prevalent disor-
der, but has the advantage of elucidating fundamental neural
Other important considerations for future research include biology capable of detecting new therapeutic targets.
inclusion of both genders in both animal and human stud- In our literature search, all human studies that involve
ies (currently most studies focus on males—see Tables 1 stimulation of deep structures were conducted exclusively

13
Deep Brain Stimulation for Addictive Disorders—Where Are We Now?

with high-frequency stimulation, as commonly used in DBS Acknowledgements  We would like to acknowledge Ms. Leslie Hassett,
for movement disorders (see Table 2). Therefore, future Outreach Librarian, Mayo Clinic, for her contribution to the thorough
literature search. M. B. is supported by a NHMRC Senior Principal
work to compare how stimulation at different frequencies Research Fellowship (1156072).
may affect the biomarkers such as LFP in humans would
be beneficial. Required Author Forms   23 provided by the authors are available with
The duration of trials may also be important. One of the the online version of this article.
possible reasons attributed to the failure of early trials in
DBS for depression is the short treatment duration, as trials Author contribution  K. H. L., Y. O., J. H. K., and J. Y. conceptualized
the study. J. Y. collected the data. J. Y. drafted the first manuscript.
with longer duration appeared to show significant efficacy A. Z. K., M. B., J. H. K., S. T., C. D. B., K. E. B., K. H. L., and Y. O.
[6, 133–135]. DBS duration effects will need to be assessed critically reviewed and revised the manuscript. K. H. L., M. B., J. H.
in larger clinical trials. Further, for neuropsychiatric condi- K., H. S., and Y. O. supervised all aspects of this work. J. Y. drafted
tions such as addiction, careful consideration of the ethical the figures. All authors accepted the final version of the manuscript.
aspect is required.
Funding  Open Access funding enabled and organized by CAUL and
its Member Institutions. This research was supported by the NIH
Ethical Considerations (R01NS112176) and the Minnesota Partnership for Biotechnology
and Medical Genomics (MNP #19.13).
Although DBS is much safer than early forms of psychosur-
gery such as lobotomy, it is not a risk-free treatment [136]. Declarations 
Therefore, clinicians are obliged to always have the patients’
best interest in their mind. The risks and benefits of the treat- Conflict of Interest  The authors declare no competing interests.
ment need to be balanced appropriately. One of the main
Open Access  This article is licensed under a Creative Commons Attri-
issues with human trials is the principle of autonomy, as bution 4.0 International License, which permits use, sharing, adapta-
patients with addictive disorders may not have the capacity tion, distribution and reproduction in any medium or format, as long
to consent to such process. Furthermore, there are concerns as you give appropriate credit to the original author(s) and the source,
that DBS (and indeed any forms of psychosurgery) may alter provide a link to the Creative Commons licence, and indicate if changes
were made. The images or other third party material in this article are
a patient’s personality, which can lead to actions and con- included in the article's Creative Commons licence, unless indicated
sequences that may otherwise not be performed [137–139]. otherwise in a credit line to the material. If material is not included in
Academic societies and government institutions should also the article's Creative Commons licence and your intended use is not
provide a medico-legal framework to ensure both medical permitted by statutory regulation or exceeds the permitted use, you will
need to obtain permission directly from the copyright holder. To view a
professionals and patients are protected. A full ethical dis- copy of this licence, visit http://​creat​iveco​mmons.​org/​licen​ses/​by/4.​0/.
cussion is out of the scope of this review. Nevertheless, we
recommend that the clinical team be multidisciplinary, with
access to the resources and expertise to assess and manage
the patients before and after the DBS surgery. References
Conclusions 1. Peacock A, Leung J, Larney S, Colledge S, Hickman M, Rehm
J, et al. Global statistics on alcohol, tobacco and illicit drug use:
Preclinical and clinical studies suggest DBS is a potential 2017 status report. Addiction. 2018;113(10):1905–26.
2. U.S. Department of Health and Human Services (HHS) OotSG.
treatment for refractory addictive disorders after other non- Facing Addiction in America: The Surgeon General’s Report on
invasive options fail. Here, we reviewed these studies and Alcohol, Drugs, and Health. Washington, DC: HHS; 2016. Avail-
their implications. However, given the uncertainty in its able from: https://​addic​tion.​surge​ongen​eral.​gov/​sites/​defau​lt/​files/​
mechanism of action and a lack of large scale randomized surge​on-​gener​als-​report.​pdf.
3. Brandon TH, Vidrine JI, Litvin EB. Relapse and relapse preven-
controlled trials, clinicians and scientists should be cau- tion. Annu Rev Clin Psychol. 2007;3:257–84.
tiously optimistic. We also provided recommendations on 4. Soyka M, Mutschler J. Treatment-refractory substance use disor-
the directions of future research, such as the need to iden- der: focus on alcohol, opioids, and cocaine. Prog Neuropsychop-
tify reliable biomarkers, for which LFPs in the NAc and harmacol Biol Psychiatry. 2016;70:148–61.
5. Edwards CA, Kouzani A, Lee KH, Ross EK. Neurostimulation
in vivo electrochemical measurement of monoamines so far devices for the treatment of neurologic disorders. Mayo Clin
appear the most promising options. With further research, Proc. 2017;92(9):1427–44.
we believe it can prove to be a highly effective treatment for 6. Yuen J, Rusheen AE, Price JB, Barath AS, Shin H, Kouzani AZ,
a selected group of patients. et al. Biomarkers for deep brain stimulation in animal models of
depression. Neuromodulation. 2021.
7. Marceglia S, Rosa M, Servello D, Porta M, Barbieri S, Moro
Supplementary Information  The online version contains supplemen- E, et al. Adaptive deep brain stimulation (aDBS) for Tourette
tary material available at https://d​ oi.o​ rg/1​ 0.1​ 007/s​ 13311-0​ 22-0​ 1229-4. syndrome. Brain Sci. 2017;8(1).

13
J. Yuen et al.

8. Greenberg BD, Malone DA, Friehs GM, Rezai AR, Kubu CS, activation in vivo depends on dopaminergic transmission. Eur J
Malloy PF, et al. Three-year outcomes in deep brain stimulation Neurosci. 2001;14(2):342–52.
for highly resistant obsessive-compulsive disorder. Neuropsy- 29. Valjent E, Pascoli V, Svenningsson P, Paul S, Enslen H, Corvol
chopharmacology. 2006;31(11):2384–93. JC, et al. Regulation of a protein phosphatase cascade allows
9. Fakhrieh-Asl G, Sadr SS, Karimian SM, Riahi E. Deep brain convergent dopamine and glutamate signals to activate ERK in
stimulation of the orbitofrontal cortex prevents the development the striatum. Proc Natl Acad Sci U S A. 2005;102(2):491–6.
and reinstatement of morphine place preference. Addiction Biol- 30. Shen W, Flajolet M, Greengard P, Surmeier DJ. Dichotomous
ogy. 2020;25(4):e12780. dopaminergic control of striatal synaptic plasticity. Science.
10. Degoulet M, Tiran-Cappello A, Combrisson E, Baunez C, Pelloux 2008;321(5890):848–51.
Y. Subthalamic low-frequency oscillations predict vulnerability to 31. Iino Y, Sawada T, Yamaguchi K, Tajiri M, Ishii S, Kasai H, et al.
cocaine addiction. Proceedings of the National Academy of Sci- Dopamine D2 receptors in discrimination learning and spine
ences of the United States of America. 2021;118(14):e2024121118. enlargement. Nature. 2020;579(7800):555–60.
11. Müller UJ, Sturm V, Voges J, Heinze HJ, Galazky I, Büntjen 32. Luscher C, Pascoli V. “Ups, downs, and sideways” of dopamine
L, et al. Nucleus accumbens deep brain stimulation for alcohol in drug addiction. Trends Neurosci. 2021;44(8):593–4.
addiction - safety and clinical long-term results of a pilot trial. 33. Creed M, Pascoli VJ, Luscher C. Addiction therapy. Refin-
Pharmacopsychiatry. 2016;49(4):170–3. ing deep brain stimulation to emulate optogenetic treatment
12. Chen L, Li N, Ge S, Lozano AM, Lee DJ, Yang C, et al. Long- of synaptic pathology. Science. 2015;347(6222):659–64.
term results after deep brain stimulation of nucleus accum- 34. Wilden JA, Qing KY, Hauser SR, McBride WJ, Irazoqui PP,
bens and the anterior limb of the internal capsule for prevent- Rodd ZA. Reduced ethanol consumption by alcohol-preferring
ing heroin relapse: an open-label pilot study. Brain Stimul. (P) rats following pharmacological silencing and deep brain
2019;12(1):175–83. stimulation of the nucleus accumbens shell. J Neurosurg.
13. Volkow ND, Morales M. The brain on drugs: from reward to 2014;120(4):997–1005.
addiction. Cell. 2015;162(4):712–25. 35. Levy D, Shabat-Simon M, Shalev U, Barnea-Ygael N, Cooper
14. Di Chiara G. Nucleus accumbens shell and core dopamine: A, Zangen A. Repeated electrical stimulation of reward-related
differential role in behavior and addiction. Behav Brain Res. brain regions affects cocaine but not “natural” reinforcement.
2002;137(1–2):75–114. J Neurosci. 2007;27(51):14179–89.
15. Howell LL, Cunningham KA. Serotonin 5-HT2 receptor inter- 36. Niu L, Guo Y, Lin Z, Shi Z, Bian T, Qi L, et al. Noninvasive ultra-
actions with dopamine function: implications for therapeutics sound deep brain stimulation of nucleus accumbens induces behav-
in cocaine use disorder. Pharmacol Rev. 2015;67(1):176–97. ioral avoidance. Science China Life sciences. 2020;63(9):1–9.
16. Muller CP, Homberg JR. The role of serotonin in drug use and 37. Wade CL, Kallupi M, Hernandez DO, Breysse E, de Guglielmo
addiction. Behav Brain Res. 2015;277:146–92. G, Crawford E, et al. High-frequency stimulation of the subtha-
17. Li Y, Simmler LD, Van Zessen R, Flakowski J, Wan JX, Deng F, lamic nucleus blocks compulsive-like re-escalation of heroin
et al. Synaptic mechanism underlying serotonin modulation of taking in rats. Neuropsychopharmacology. 2017;42(9):1850–9.
transition to cocaine addiction. Science. 2021;373(6560):1252–6. 38. OSF Home. Deep brain stimulation for addiction - a scoping
18. Samaha AN, Khoo SY, Ferrario CR, Robinson TE. Dopamine “ups and review and recommendations for future research: Center for
downs” in addiction revisited. Trends Neurosci. 2021;44(7):516–26. Open Science; 2021 [8/29/21]. Available from: https://​osf.​io/​
19. McDevitt RA, Tiran-Cappello A, Shen H, Balderas I, Britt JP, pqyzn/?​view_​only=​d6ccd​897bc​76442​b8eac​e0902​5451c​0f.
Marino RAM, et al. Serotonergic versus nonserotonergic dorsal 39. Liu H-Y, Jin J, Tang J-S, Sun W-X, Jia H, Yang X-P, et al. Chronic
raphe projection neurons: differential participation in reward cir- deep brain stimulation in the rat nucleus accumbens and its effect
cuitry. Cell Rep. 2014;8(6):1857–69. on morphine reinforcement. Addict Biol. 2008;13(1):40–6.
20. Nakamura K. The role of the dorsal raphe nucleus in reward- 40. Vassoler FM, Schmidt HD, Gerard ME, Famous KR, Ciraulo
seeking behavior. Front Integr Neurosci. 2013;7:60. DA, Kornetsky C, et al. Deep brain stimulation of the nucleus
21. Zweifel LS, Argilli E, Bonci A, Palmiter RD. Role of NMDA accumbens shell attenuates cocaine priming-induced reinstate-
receptors in dopamine neurons for plasticity and addictive behav- ment of drug seeking in rats. J Neurosci. 2008;28(35):8735–9.
iors. Neuron. 2008;59(3):486–96. 41. Guo L, Zhou H, Wang R, Xu J, Zhou W, Zhang F, et al. DBS of nucleus
22. Luscher C, Janak PH. Consolidating the circuit model for addic- accumbens on heroin seeking behaviors in self-administering rats.
tion. Annu Rev Neurosci. 2021;44:173–95. Drug Alcohol Depend. 2013;129(1–2):70–81.
23. Girault JA, Greengard P. The neurobiology of dopamine signal- 42. Vassoler FM, White SL, Hopkins TJ, Guercio LA, Espallergues
ing. Arch Neurol. 2004;61(5):641–4. J, Berton O, et al. Deep brain stimulation of the nucleus accum-
24. Yagishita S, Hayashi-Takagi A, Ellis-Davies GC, Urakubo bens shell attenuates cocaine reinstatement through local and
H, Ishii S, Kasai H. A critical time window for dopamine antidromic activation. J Neurosci. 2013;33(36):14446–54.
actions on the structural plasticity of dendritic spines. Science. 43. Hamilton J, Lee J, Canales JJ. Chronic unilateral stimulation
2014;345(6204):1616–20. of the nucleus accumbens at high or low frequencies attenuates
25. Brown RML, A. J.; Kim, J. H. The role of mitogen-activated relapse to cocaine seeking in an animal model. Brain Stimul.
protein kinase in treatment strategies for fear and drug addiction. 2015;8(1):57–63.
Advances in Protein Kinases. London: IntechOpen; 2012. 44. Hadar R, Vengeliene V, Barroeta Hlusicke E, Canals S, Noori
26. Bertran-Gonzalez J, Bosch C, Maroteaux M, Matamales M, HR, Wieske F, et al. Paradoxical augmented relapse in alcohol-
Herve D, Valjent E, et al. Opposing patterns of signaling acti- dependent rats during deep-brain stimulation in the nucleus
vation in dopamine D1 and D2 receptor-expressing striatal accumbens. Transl Psychiatry Psychiatry. 2016;6(6):e840.
neurons in response to cocaine and haloperidol. J Neurosci. 45. Batra V, Tran TLN, Caputo J, Guerin GF, Goeders NE, Wilden
2008;28(22):5671–85. J. Intermittent bilateral deep brain stimulation of the nucleus
27. Valjent E, Corvol JC, Trzaskos JM, Girault JA, Herve D. Role of accumbens shell reduces intravenous methamphetamine intake
the ERK pathway in psychostimulant-induced locomotor sensi- and seeking in Wistar rats. J Neurosurg. 2017;126(4):1339–50.
tization. BMC Neurosci. 2006;7:20. 46. Mehdipour S, Alaei HA, Pilehvariyan AA. The effect of medial
28. Valjent E, Pages C, Rogard M, Besson MJ, Maldonado R, Caboche prefrontal cortex electrical stimulation on passive avoidance
J. Delta 9-tetrahydrocannabinol-induced MAPK/ERK and Elk-1 memory in healthy and addict rats. Adv. 2015;4:254.

13
Deep Brain Stimulation for Addictive Disorders—Where Are We Now?

47. Guercio LA, Wimmer ME, Swinford-Jackson SE, Pierce RC, shell dissociates encoding of values for reward and pain. J Neu-
Vassoler FM, Schmidt HD. Deep brain stimulation of the infral- rosci. 2013;33(41):16383–93.
imbic cortex attenuates cocaine priming-induced reinstatement 65. Budilin SY, Midzyanovskaya IS, Shchegolevskii NV, Ioffe
of drug seeking. Brain Research. 2020;1746:147011. ME, Bazyan AS. Asymmetry in dopamine levels in the nucleus
48. Scofield MD, Heinsbroek JA, Gipson CD, Kupchik YM, Spencer S, accumbens and motor preference in rats. Neurosci Behav Physiol.
Smith AC, et al. The nucleus accumbens: mechanisms of addiction 2008;38(9):991–4.
across drug classes reflect the importance of glutamate homeostasis. 66. Navarro PA, Lopez WOC, Paranhos T, Lovo E, De Oliveira-
Pharmacol Rev. 2016;68(3):816–71. Souza R, Gorgulho AA, et al. Safety and feasibility of nucleus
49. Salgado S, Kaplitt MG. The nucleus accumbens: a comprehen- accumbens surgery for drug addiction: a systematic review. Neu-
sive review. Stereotact Funct Neurosurg. 2015;93(2):75–93. romodulation. 2021.
50. Gibson GD, Millan EZ, McNally GP. The nucleus accumbens 67. Kuhn J, Moller M, Treppmann JF, Bartsch C, Lenartz D,
shell in reinstatement and extinction of drug seeking. Eur J Neu- Gruendler TOJ, et al. Deep brain stimulation of the nucleus
rosci. 2019;50(3):2014–22. accumbens and its usefulness in severe opioid addiction. Mol
51. Knapp CM, Tozier L, Pak A, Ciraulo DA, Kornetsky C. Deep Psychiatry. 2014;19(2):145–6.
brain stimulation of the nucleus accumbens reduces ethanol con- 68. Zhu R, Zhang Y, Wang T, Wei H, Zhang C, Li D, et al. Deep
sumption in rats. Pharmacol Biochem Behav. 2009;92(3):474–9. brain stimulation of nucleus accumbens with anterior capsul-
52. Henderson MB, Green AI, Bradford PS, Chau DT, Roberts DW, otomy for drug addiction: a case report. Stereotact Funct Neuro-
Leiter JC. Deep brain stimulation of the nucleus accumbens surg. 2020;98(5):345–9.
reduces alcohol intake in alcohol-preferring rats. Neurosurg 69. Hill DF, Parent KL, Atcherley CW, Cowen SL, Heien ML. Dif-
Focus. 2010;29(2):E12. ferential release of dopamine in the nucleus accumbens evoked
53. Pushparaj A, Hamani C, Yu W, Shin DS, Kang B, Nobrega by low-versus high-frequency medial prefrontal cortex stimula-
JN, et al. Electrical stimulation of the insular region attenuates tion. Brain Stimul. 2018;11(2):426–34.
nicotine-taking and nicotine-seeking behaviors. Neuropsychop- 70. Zbukvic IC, Kim JH. Divergent prefrontal dopaminergic mecha-
harmacology. 2013;38(4):690–8. nisms mediate drug- and fear-associated cue extinction during
54. Beaulieu CL, Thorn BE. Focal brain stimulation attenu- adolescence versus adulthood. Eur Neuropsychopharmacol.
ates morphine withdrawal behaviors. Behav Neurosci. 2018;28(1):1–12.
1986;100(4):504–11. 71. Lupi M, Sepede G, Cinosi E, Martinotti G, di Giannantonio M.
55. Ma Y, Chen N, Wang HM, Meng FG, Zhang JG. Inhibition of the The efficacy of transcranical direct current stimulation in prega-
reinstatement of morphine-induced place preference in rats by balin abuse: a case report. Journal of ECT. 2018;34(1):e14–5.
high-frequency stimulation of the bilateral nucleus accumbens. 72. Perini I, Kampe R, Arlestig T, Karlsson H, Lofberg A, Pietrzak
Chin Med J (Engl). 2013;126(10):1939–43. M, et al. Repetitive transcranial magnetic stimulation targeting
56. Martínez-Rivera FJ, Rodriguez-Romaguera J, Lloret-Torres ME, the insular cortex for reduction of heavy drinking in treatment-
Do Monte FH, Quirk GJ, Barreto-Estrada JL. Bidirectional mod- seeking alcohol-dependent subjects: a randomized controlled trial.
ulation of extinction of drug seeking by deep brain stimulation Neuropsychopharmacology : official publication of the American
of the ventral striatum. Biol Psychiatry. 2016;80(9):682–90. College of Neuropsychopharmacology. 2020;45(5):842–50.
57. Chang H, Gao C, Sun K, Xiao L, Li X, Jiang S, et al. Continuous 73. Ridderinkhof KR, de Vlugt Y, Bramlage A, Spaan M,
high frequency deep brain stimulation of the rat anterior insula Elton M, Snel J, et  al. Alcohol consumption impairs detec-
attenuates the relapse post withdrawal and strengthens the extinc- tion of performance errors in mediofrontal cortex. Science.
tion of morphine seeking. Frontiers in Psychiatry. 2020;11 (no 2002;298(5601):2209–11.
pagination). 74. Schellekens AF, de Bruijn ER, van Lankveld CA, Hulstijn W, Buitelaar
58. Friedman A, Lax E, Dikshtein Y, Abraham L, Flaumenhaft Y, JK, de Jong CA, et al. Alcohol dependence and anxiety increase
Sudai E, et al. Electrical stimulation of the lateral habenula pro- error-related brain activity. Addiction. 2010;105(11):1928–34.
duces enduring inhibitory effect on cocaine seeking behavior. 75. Leong SL, Glue P, Manning P, Vanneste S, Lim LJ, Mohan A,
Neuropharmacology. 2010;59(6):452–9. et al. Anterior cingulate cortex implants for alcohol addiction: a
59. Rouaud T, Lardeux S, Panayotis N, Paleressompoulle D, Cador feasibility study. Neurotherapeutics. 2020;17(3):1287–99.
M, Baunez C. Reducing the desire for cocaine with subthalamic 76. De Ridder D, Vanneste S, Kovacs S, Sunaert S, Dom G. Transient
nucleus deep brain stimulation. Proc Natl Acad Sci U S A. alcohol craving suppression by rTMS of dorsal anterior cingulate: an
2010;107(3):1196–200. fMRI and LORETA EEG study. Neurosci Lett. 2011;496(1):5–10.
60. Maks CB, Butson CR, Walter BL, Vitek JL, McIntyre CC. Deep 77. De Ridder D, Leong SL, Manning P, Vanneste S, Glue P. Ante-
brain stimulation activation volumes and their association with rior cingulate implant for obsessive-compulsive disorder. World
neurophysiological mapping and therapeutic outcomes. J Neurol Neurosurgery. 2017;97:754.e7-.e16.
Neurosurg Psychiatry. 2009;80(6):659–66. 78. Kuhn J, Gründler TO, Bauer R, Huff W, Fischer AG, Lenartz D,
61. Baumann B, Danos P, Krell D, Diekmann S, Leschinger A, et al. Successful deep brain stimulation of the nucleus accumbens
Stauch R, et al. Reduced volume of limbic system-affiliated basal in severe alcohol dependence is associated with changed perfor-
ganglia in mood disorders: preliminary data from a postmortem mance monitoring. Addict Biol. 2011;16(4):620–3.
study. J Neuropsychiatry Clin Neurosci. 1999;11(1):71–8. 79. Ibrahim C, Rubin-Kahana DS, Pushparaj A, Musiol M, Blumberger
62. Wong JE, Cao J, Dorris DM, Meitzen J. Genetic sex and the volumes DM, Daskalakis ZJ, et al. The insula: a brain stimulation target for
of the caudate-putamen, nucleus accumbens core and shell: original the treatment of addiction. Front Pharmacol. 2019;10:720.
data and a review. Brain Struct Funct. 2016;221(8):4257–67. 80. Kalivas PW, Volkow ND. The neural basis of addiction: a pathology
63. Xia X, Fan L, Cheng C, Eickhoff SB, Chen J, Li H, et al. Mul- of motivation and choice. Am J Psychiatry. 2005;162(8):1403–13.
timodal connectivity-based parcellation reveals a shell-core 81. Peters J, Kalivas PW, Quirk GJ. Extinction circuits for fear and addic-
dichotomy of the human nucleus accumbens. Hum Brain Mapp. tion overlap in prefrontal cortex. Learn Mem. 2009;16(5):279–88.
2017;38(8):3878–98. 82. Witjas T, Baunez C, Henry JM, Delfini M, Regis J, Cherif AA,
64. Baliki MN, Mansour A, Baria AT, Huang L, Berger SE, Fields et al. Addiction in Parkinson’s disease: impact of subthalamic
HL, et al. Parceling human accumbens into putative core and nucleus deep brain stimulation. Mov Disord. 2005;20(8):1052–5.

13
J. Yuen et al.

83. Eusebio A, Witjas T, Cohen J, Fluchere F, Jouve E, Regis J, et al. 102. Boutet A, Ranjan M, Zhong J, Germann J, Xu D, Schwartz
Subthalamic nucleus stimulation and compulsive use of dopa- ML, et  al. Focused ultrasound thalamotomy location deter-
minergic medication in Parkinson’s disease. J Neurol Neurosurg mines clinical benefits in patients with essential tremor. Brain.
Psychiatry. 2013;84(8):868–74. 2018;141(12):3405–14.
84. Baunez C, Dias C, Cador M, Amalric M. The subthalamic 103. Davidson B, Hamani C, Huang Y, Jones RM, Meng Y, Giacobbe
nucleus exerts opposite control on cocaine and “natural” rewards. P, et al. Magnetic resonance-guided focused ultrasound capsu-
Nat Neurosci. 2005;8(4):484–9. lotomy for treatment-resistant psychiatric disorders. Oper Neu-
85. Pinel JP, Van Oot PH, Mucha RF. Intensification of the alco- rosurg (Hagerstown). 2020;19(6):741–9.
hol withdrawal syndrome by repeated brain stimulation. Nature. 104. Gao G, Wang X, He S, Li W, Wang Q, Liang Q, et al. Clinical
1975;254(5500):510–1. study for alleviating opiate drug psychological dependence by
86. Lozano AM, Lipsman N, Bergman H, Brown P, Chabardes S, a method of ablating the nucleus accumbens with stereotactic
Chang JW, et al. Deep brain stimulation: current challenges and surgery. Stereotact Funct Neurosurg. 2003;81(1–4):96–104.
future directions. Nat Rev Neurol. 2019;15(3):148–60. 105. Hariz MI, Shamsgovara P, Johansson F, Hariz G, Fodstad H.
87. McCracken CB, Grace AA. High-frequency deep brain stimula- Tolerance and tremor rebound following long-term chronic tha-
tion of the nucleus accumbens region suppresses neuronal activ- lamic stimulation for Parkinsonian and essential tremor. Stereo-
ity and selectively modulates afferent drive in rat orbitofrontal tact Funct Neurosurg. 1999;72(2–4):208–18.
cortex in vivo. J Neurosci. 2007;27(46):12601–10. 106. McCracken CB, Grace AA. Nucleus accumbens deep brain
88. Wise RA. Addictive drugs and brain stimulation reward. Annu stimulation produces region-specific alterations in local field
Rev Neurosci. 1996;19:319–40. potential oscillations and evoked responses in vivo. J Neurosci.
89. Bauer CT, Banks ML, Blough BE, Negus SS. Use of intracranial self- 2009;29(16):5354–63.
stimulation to evaluate abuse-related and abuse-limiting effects of 107. Ge S, Geng X, Wang X, Li N, Chen L, Zhang X, et al. Oscillatory
monoamine releasers in rats. Br J Pharmacol. 2013;168(4):850–62. local field potentials of the nucleus accumbens and the anterior
90. Zhou H, Xu J, Jiang J. Deep brain stimulation of nucleus accum- limb of the internal capsule in heroin addicts. Clin Neurophysiol.
bens on heroin-seeking behaviors: a case report. Biol Psychiat. 2018;129(6):1242–53.
2011;69(11):e41–2. 108. Stenner MP, Durschmid S, Rutledge RB, Zaehle T, Schmitt
91. Zhang C, Li J, Li D, Sun B. Deep brain stimulation removal after FC, Kaufmann J, et al. Perimovement decrease of alpha/beta
successful treatment for heroin addiction. Aust N Z J Psychiatry. oscillations in the human nucleus accumbens. J Neurophysiol.
2020;54(5):543–4. 2016;116(4):1663–72.
92. Heldmann M, Berding G, Voges J, Bogerts B, Galazky I, Müller U, 109. Miller KJ, Prieto T, Williams NR, Halpern CH. Case studies
et al. Deep brain stimulation of nucleus accumbens region in alco- in neuroscience: the electrophysiology of a human obsession in
holism affects reward processing. PLoS One. 2012;7(5):e36572. nucleus accumbens. J Neurophysiol. 2019;121(6):2336–40.
93. Kuhn J, Bauer R, Pohl S, Lenartz D, Huff W, Kim EH, et al. Obser- 1 10. Medtronic. PERCEPT PC NEUROSTIMULATOR 2020
vations on unaided smoking cessation after deep brain stimulation [Available from: https:// ​ w ww. ​ m edtr ​ o nic. ​ c om/ ​ u k- ​ e n/​
of the nucleus accumbens. Eur Addict Res. 2009;15(4):196–201. patie ​ n ts/ ​ t reat ​ m ents- ​ t hera ​ p ies/ ​ d eep- ​ b rain- ​ s timu ​ l ation-
94. Mantione M, van de Brink W, Schuurman PR, Denys D. Smoking ​p arki​n sons-​d isea​s e/​w hy-​choose-​m edtr​o nic/​o ur-​m edtr​o nic-​
cessation and weight loss after chronic deep brain stimulation of dbs-​s ystem.​h tml.
the nucleus accumbens: therapeutic and research implications: 111. U.S. National Library of Medicine. Brain electrophysiological
case report. Neurosurgery. 2010;66(1):E218; discussion E. study(EEG/ERP) on opiate addicts treating by bilateral NAc/
95. Lupi M, Martinotti G, Santacroce R, Cinosi E, Carlucci M, ALIC deep brain stimulation 2021 [6/6/21]. Available from:
Marini S, et al. Transcranial direct current stimulation in sub- https://​clini​caltr​ials.​gov/​ct2/​show/​NCT02​594306?​term=​deep+​
stance use disorders: a systematic review of scientific literature. brain+​stimu​latio​n&​cond=​Addic​tion&​draw=​2&​rank=​13.
Journal of ECT. 2017;33(3):203–9. 112. Chefer VI, Thompson AC, Zapata A, Shippenberg TS. Overview of
96. Ekhtiari H, Tavakoli H, Addolorato G, Baeken C, Bonci A, brain microdialysis. Curr Protoc Neurosci. 2009;Chapter 7:Unit7 1.
Campanella S, et al. Transcranial electrical and magnetic stimu- 113. Robinson DL, Venton BJ, Heien ML, Wightman RM. Detecting
lation (tES and TMS) for addiction medicine: a consensus paper subsecond dopamine release with fast-scan cyclic voltammetry
on the present state of the science and the road ahead. Neurosci in vivo. Clin Chem. 2003;49(10):1763–73.
Biobehav Rev. 2019;104:118–40. 114. Heien ML, Johnson MA, Wightman RM. Resolving neurotrans-
97. Diana M, Raij T, Melis M, Nummenmaa A, Leggio L, Bonci mitters detected by fast-scan cyclic voltammetry. Anal Chem.
A. Rehabilitating the addicted brain with transcranial magnetic 2004;76(19):5697–704.
stimulation. Nat Rev Neurosci. 2017;18(11):685–93. 115. Rodeberg NT, Sandberg SG, Johnson JA, Phillips PEM, Wightman
98. Hone-Blanchet A, Ciraulo DA, Pascual-Leone A, Fecteau S. Non- RM. Hitchhiker’s guide to voltammetry: acute and chronic elec-
invasive brain stimulation to suppress craving in substance use dis- trodes for in vivo fast-scan cyclic voltammetry. ACS Chem Neuro-
orders: review of human evidence and methodological considera- sci. 2017;8:221−34.
tions for future work. Neurosci Biobehav Rev. 2015;59:184–200. 116. Clark JJ, Sandberg SG, Wanat MJ, Gan JO, Horne EA, Hart AS,
99. Ward HB, Mosquera MJ, Suzuki J, Mariano TY. A systematic et al. Chronic microsensors for longitudinal, subsecond dopamine
review of noninvasive brain stimulation for opioid use disorder. detection in behaving animals. Nat Methods. 2010;7(2):126–9.
Neuromodulation. 2020;23(3):301–11. 117. Yuen J, Goyal A, Rusheen AE, Kouzani AZ, Berk M, Kim JH,
100. Jansen JM, Daams JG, Koeter MWJ, Veltman DJ, van den Brink W, et al. Cocaine increases stimulation-evoked serotonin efflux in
Goudriaan AE. Effects of non-invasive neurostimulation on craving: the nucleus accumbens. J Neurophysiol. 2022.
a meta-analysis. Neurosci Biobehav Rev. 2013;37(10 Pt 2):2472–80. 118. Yuen J, Goyal A, Rusheen AE, Kouzani AZ, Berk M, Kim JH,
101. Abbott. Abbott introduces neurosphere™ virtual clinic, first- et al. Cocaine-induced changes in tonic dopamine concentra-
of-its-kind remote neuromodulation patient-care technology tions measured using multiple-cyclic square wave voltammetry
in the U.S. 2021 [Available from: https://​abbott.​media​room.​ in vivo. Frontiers in Pharmacology. 2021;12(1710).
com/​2021-​03-​08-​Abbott-​Intro​duces-​Neuro​Sphere-​TM-​Virtu​al-​ 119. Oh Y, Heien ML, Park C, Kang YM, Kim J, Boschen SL, et al.
Clinic-F​ irst-o​ f-i​ ts-K
​ ind-R
​ emote-N
​ eurom
​ odula​ tion-P
​ atien​ t-C
​ are-​ Tracking tonic dopamine levels in vivo using multiple cyclic square
Techn​ology-​in-​the-U-S. wave voltammetry. Biosens Bioelectron. 2018;121:174–82.

13
Deep Brain Stimulation for Addictive Disorders—Where Are We Now?

120. Atcherley CW, Wood KM, Parent KL, Hashemi P, Heien ML. 136. Caruso JP, Sheehan JP. Psychosurgery, ethics, and media: a his-
The coaction of tonic and phasic dopamine dynamics. Chem tory of Walter Freeman and the lobotomy. Neurosurg Focus.
Commun (Camb). 2015;51(12):2235–8. 2017;43(3):E6.
121. Rusheen AE, Gee TA, Jang DP, Blaha CD, Bennet KE, Lee KH, 137. Freeman W. Ethics of psychosurgery. N Engl J Med.
et al. Evaluation of electrochemical methods for tonic dopamine 1953;249(20):798–801.
detection in vivo. Trends Analyt Chem. 2020;132. 138. Ali R, DiFrancesco MF, Ho AL, Kampman KM, Caplan AL,
122. Shin H, Goyal A, Barnett JH, Rusheen AE, Yuen J, Jha R, et al. Halpern CH. Attitudes toward treating addiction with deep brain
Tonic serotonin measurements in vivo using N-shaped multiple stimulation. Brain Stimul. 2016;9(3):466–8.
cyclic square wave voltammetry. Anal Chem. 2021. 139. Trujols J, Manresa MJ, Batlle F, Duran-Sindreu S, Pérez de Los
123. Gittis AH, Yttri EA. Translating insights from optogenetics Cobos J. Deep brain stimulation for addiction treatment: further
to therapies for Parkinson’s disease. Curr Opin Biomed Eng. considerations on scientific and ethical issues. Brain Stimul.
2018;8:14–9. 2016;9(5):788–9.
124. Spix TA, Nanivadekar S, Toong N, Kaplow IM, Isett BR, 140. Volkow ND, Koob GF, McLellan AT. Neurobiologic advances
Goksen Y, et  al. Population-specific neuromodulation pro- from the brain disease model of addiction. N Engl J Med.
longs therapeutic benefits of deep brain stimulation. Science. 2016;374(4):363–71.
2021;374(6564):201–6. 141. Kuhn J, Lenartz D, Huff W, Lee S, Koulousakis A, Klosterkoetter
125. Schor JS, Montalvo IG, Spratt PWE, Brakaj RJ, Stansil JA, J, et al. Remission of alcohol dependency following deep brain
Bender KJ, et al. Therapeutic deep brain stimulation disrupts stimulation of the nucleus accumbens: valuable therapeutic
subthalamic nucleus activity dynamics in Parkinsonian mice implications? Journal of Neurology, Neurosurgery & Psychiatry.
(Preprint). bioRxiv. 2021:2021.11.12.468404. 2007;78(10):1152–3.
126. Patel AA, McAlinden N, Mathieson K, Sakata S. Simultaneous 142. Valencia-Alfonso CE, Luigjes J, Smolders R, Cohen MX, Levar
electrophysiology and fiber photometry in freely behaving mice. N, Mazaheri A, et al. Effective deep brain stimulation in heroin
Front Neurosci. 2020;14:148. addiction: a case report with complementary intracranial elec-
127. U.S. National Library of Medicine. ClinicalTrials.gov 2021 troencephalogram. Biol Psychiatry. 2012;71(8):e35–7.
[6/6/21]. Available from: https://​clini​caltr​ials.​gov/​ct2/​home. 143. Voges J, Muller U, Bogerts B, Munte T, Heinze H-J. Deep brain
128. Goldstein RZ, Volkow ND. Dysfunction of the prefrontal cortex stimulation surgery foralcohol addiction. World Neurosurgery.
in addiction: neuroimaging findings and clinical implications. 2013;80(3-4):S28.e1–31.
Nat Rev Neurosci. 2011;12(11):652–69. 144. Muller UJ, Sturm V, Voges J, Heinze HJ, Galazky I, Heldmann
129. Formolo DA, Gaspar JM, Melo HM, Eichwald T, Zepeda RJ, M, et al. Successful treatment of chronic resistant alcoholism by
Latini A, et al. Deep brain stimulation for obesity: a review and deep brain stimulation of nucleus accumbens: first experience
future directions. Front Neurosci. 2019;13:323. with three cases. Pharmacopsychiatry. 2009;42(6):288–91.
130. Casquero-Veiga M, Bueno-Fernandez C, Romero-Miguel D, 145. Heinze HJ, Heldmann M, Voges J, Hinrichs H, Marco-Pallares
Lamanna-Rama N, Nacher J, Desco M, et al. Exploratory study J, Hopf JM, et al. Counteracting incentive sensitization in severe
of the long-term footprint of deep brain stimulation on brain alcohol dependence using deep brain stimulation of the nucleus
metabolism and neuroplasticity in an animal model of obesity. accumbens: clinical and basic science aspects. Front Hum Neu-
Sci Rep. 2021;11(1):5580. rosci. 2009;3:22.
131. Halpern CH, Torres N, Hurtig HI, Wolf JA, Stephen J, Oh MY, 146. Gonçalves-Ferreira A, do Couto FS, Rainha Campos A, Lucas
et al. Expanding applications of deep brain stimulation: a poten- Neto LP, Gonçalves-Ferreira D, Teixeira J. Deep Brain Stimu-
tial therapeutic role in obesity and addiction management. Acta lation for Refractory Cocaine Dependence. Biol Psychiatry.
Neurochir (Wien). 2011;153(12):2293–306. 2016;79(11):e87–9.
132. Zhang C, Huang Y, Zheng F, Zeljic K, Pan J, Sun B. Death from 147. Ge S, Chen Y, Li N, Qu L, Li Y, Jing J, et al. Deep Brain Stimu-
opioid overdose after deep brain stimulation: a case report. Biol lation of Nucleus Accumbens for Methamphetamine Addiction:
Psychiatry. 2018;83(1):e9–10. Two Case Reports. World Neurosurg. 2019;122:512–7.
133. Dougherty DD, Rezai AR, Carpenter LL, Howland RH, Bhati 148. Sildatke E, Schuller T, Huys D, Grundler TOJ, Ullsperger M,
MT, O’Reardon JP, et al. A randomized sham-controlled trial Visser-Vandewalle V, et al. Error-Related Activity in Striatal
of deep brain stimulation of the ventral capsule/ventral stria- Local Field Potentials and Medial Frontal Cortex: Evidence
tum for chronic treatment-resistant depression. Biol Psychiatry. From Patients With Severe Opioid Abuse Disorder. Frontiers in
2015;78(4):240–8. Human Neuroscience. 2020;14:627564.
134. Crowell AL, Riva-Posse P, Holtzheimer PE, Garlow SJ, Kelley 149. Mahoney JJ, Haut MW, Hodder SL, Zheng W, Lander LR, Berry
ME, Gross RE, et al. Long-term outcomes of subcallosal cingu- JH, et al. Deep brain stimulation of the nucleus accumbens/ven-
late deep brain stimulation for treatment-resistant depression. Am tral capsule for severe and intractable opioid and benzodiazepine
J Psychiatry. 2019;176(11):949–56. use disorder. Experimental and clinical psychopharmacology.
135. Holtzheimer PE, Husain MM, Lisanby SH, Taylor SF, Whitworth 2021;29(2):210–5.
LA, McClintock S, et al. Subcallosal cingulate deep brain stimu-
lation for treatment-resistant depression: a multisite, randomised, Publisher's Note Springer Nature remains neutral with regard to
sham-controlled trial. Lancet Psychiatry. 2017;4(11):839–49. jurisdictional claims in published maps and institutional affiliations.

13

You might also like