Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

REVIEWS

Technology of deep brain stimulation:


current status and future directions
Joachim K. Krauss 1, Nir Lipsman 2, Tipu Aziz3, Alexandre Boutet 4, Peter Brown 5
,
Jin Woo Chang6, Benjamin Davidson 2, Warren M. Grill 7, Marwan I. Hariz8,
Andreas Horn 9, Michael Schulder10, Antonios Mammis11, Peter A. Tass 12,
Jens Volkmann1,13 and Andres M. Lozano 14 ✉
Abstract | Deep brain stimulation (DBS) is a neurosurgical procedure that allows targeted
circuit-​based neuromodulation. DBS is a standard of care in Parkinson disease, essential tremor
and dystonia, and is also under active investigation for other conditions linked to pathological
circuitry, including major depressive disorder and Alzheimer disease. Modern DBS systems,
borrowed from the cardiac field, consist of an intracranial electrode, an extension wire and a pulse
generator, and have evolved slowly over the past two decades. Advances in engineering and
imaging along with an improved understanding of brain disorders are poised to reshape how DBS
is viewed and delivered to patients. Breakthroughs in electrode and battery designs, stimulation
paradigms, closed-​loop and on-​demand stimulation, and sensing technologies are expected to
enhance the efficacy and tolerability of DBS. In this Review, we provide a comprehensive overview
of the technical development of DBS, from its origins to its future. Understanding the evolution of
DBS technology helps put the currently available systems in perspective and allows us to predict
the next major technological advances and hurdles in the field.

The opportunities to use technology to modulate or potential uses and prompted studies on novel applica-
influence brain circuitry and human behaviour have tions for conditions such as tinnitus, arterial hyperten-
increased exponentially over the past few years. These sion and sleep disorders9. Chronic stimulation not only
developments have spawned the field of electroceuti- has direct physiological effects on brain circuits but also
cals, with deep brain stimulation (DBS) being the most produces a range of cellular, molecular and neuroplastic
important and accepted treatment within this class of changes1. Our increased understanding of the complex
therapies1. DBS is commonly indicated for the treatment mode of action of DBS is leading to a more comprehen-
of movement disorders such as Parkinson disease (PD), sive appreciation of the effects of chronic stimulation in
tremor and dystonia, and became the standard of care the nervous system.
for these conditions after receiving FDA and Conformité The technology for DBS was developed by mod-
Européene (CE) approval2–4. In addition, DBS has been ifying cardiac pacemakers and saw little development
used for pain syndromes, such as neuropathic pain and or advancement for almost two decades following the
cluster headache, as well as for epilepsy5–7. A favourable inception of the ‘modern’ DBS era in the late 1980s
safety profile and demonstration of efficacy in several (Fig. 1). Until recently, technological advancements in the
randomized controlled trials has led to increased inter- field have been driven largely by limitations of DBS tech-
est in the potential application of DBS to psychiatric nology such as large battery size, limited battery life and
disorders1. Following a positive randomized controlled the need for frequent battery replacements. However, the
trial published in 2008, DBS for obsessive–compulsive appearance of multiple manufacturers of DBS technol-
disorder (OCD) was granted CE approval and an FDA ogy on the global market has sparked international com-
Humanitarian Device Exemption8,9, and DBS is cur- petition and we are now seeing progress at an accelerated
rently under investigation for a wide range of other pace. In the coming years, we anticipate the implementa-
treatment-​resistant conditions, including depression, tion of new hardware designs, improved technology and
✉e-​mail: andres.lozano@ Alzheimer disease, Tourette syndrome, addiction, ano- refined stimulation algorithms. Advances in DBS tech-
uhnresearch.ca rexia nervosa and schizophrenia9. The minimally inva- nology will no doubt extend the scope of its application
https://doi.org/10.1038/ sive character of DBS, combined with the low incidence and are expected to yield additional benefits, both clini-
s41582-020-00426-​z of severe, disabling adverse effects, has expanded its cally and scientifically10–12. We expect these advances to

Nature Reviews | Neurology


Reviews

History of DBS technical innovation


Key points
The history of DBS began with its use in psychiatric
• Deep brain stimulation (DBS) is a neurosurgical procedure that allows targeted disease and pain15. The use of subcortically implanted
circuit-​based neuromodulation and is commonly used for the treatment of movement electrodes for therapeutic chronic stimulation was
disorders such as Parkinson disease, tremor and dystonia. first described by Columbia University neurosurgeon
• Innovations in the field of cardiac pacemakers have enabled pulse generators for DBS Lawrence Pool. In 1948, Pool implanted an electrode into
to evolve from external devices to small rechargeable, implantable devices. the head of the caudate nucleus in a woman with depres-
• With directional DBS leads, the current can be directed or shaped to personalize sion and anorexia and reported “favourable results” for
stimulation to individual anatomical structures. several weeks until the wire broke16. In 1952, Yale neuro-
• Closed-​loop DBS systems simultaneously record and stimulate neural activity, physiologist José Delgado and colleagues started a pro-
allowing the stimulation to be adjusted according to disease-​specific neural gramme of chronic stimulation of deep brain structures
biomarkers. in patients with psychiatric illnesses17. With remarkable
• Open-​access software can be used to localize DBS electrodes and, on the basis of the prescience, they invented a so-​called ‘stimoceiver’ that
stimulation parameters, to model the volume of tissue activated around the was implanted in the skull, thereby allowing remote acti-
electrodes, shedding light on key neurocircuitry elements.
vation of the stimulator18. Following the identification of
• As DBS systems become compatible with wireless networks, remote programming by ‘pleasure’ brain targets associated with self-​stimulation
physicians will become possible but privacy issues will also need to be addressed to
via intracranial electrodes in experimental animals
prevent misuse, including ‘brainjacking’.
at McGill University, controversial Tulane University
psychiatrist Robert Heath developed the technique of
Brainjacking lead to enhanced market penetration and a wider acces- high-​frequency (100 Hz) chronic stimulation, which he
The unauthorized control of an sibility of DBS to the populations and patients who can subsequently applied in the septal area of the brain to
implanted brain device, benefit the most, including those in low-​income nations. treat schizophrenia and pain19,20.
theoretically through Bluetooth However, it is important to be aware of new dangers that Norwegian neurophysiologist and psychiatrist Carl
or wireless internet technology.
might arise with advances in electronics and comput- Wilhelm Sem-​Jacobsen and colleagues at the Mayo
Gate theory ing, such as the prospect of modulation of cognitive Clinic contributed to the development of chronic
Theory describing the ‘gating’ and decision-​making processes, and the possibility of subcortical stimulation with the intent of finding the
of pain signals, whereby the acquiring data for misuse and brainjacking13,14. best sites for subsequent neural ablation in patients
transmission of non-​painful
In this article, we review the evolution and current with psychiatric disorders21. On returning to Oslo,
stimuli can block or override
painful signals at the level of
status of DBS technology, anticipate future advances Sem-​Jacobsen continued this technique and extended
the spinal cord. and discuss their clinical implications. We provide a rea- it to the treatment of patients with PD22. Again, the
soned overview of new DBS electrodes and pulse gener- aim was to apply chronic stimulation over several
ators as well as of innovations in stimulation algorithms weeks to identify the optimal ablation target. In par-
and programming. We also discuss developments in the allel, Leningrad neurophysiologists Natalia Bechtereva
imaging of implanted electrodes, which has become rel- and colleagues used a similar technique termed ther-
evant not only for practical reasons but also in under- apeutic electrostimulation23,24. Chronic stimulation
standing brain dysfunction and enhancing the efficacy was delivered repetitively over weeks or months and
of chronic neurostimulation. Finally, we address the eth- a lesion was eventually made at the site where stim-
ical and security issues raised by new technical develop- ulation yielded the best clinical results. The use of
ments, in particular with regard to potential scenarios chronic stimulation without subsequent lesioning only
of misuse, including by third parties. We conclude by became possible with the introduction of reliable pulse
predicting future directions for the field of DBS technol- generators25.
ogy and consider how changes in DBS will enhance the The development of neurostimulation owes much
accessibility of this effective surgical technique to those to the battery and implant design of cardiac pacemaker
patients who are most in need. technology. Following the publication of the gate theory
by Ronald Melzack and Patrick Wall in 1965 (ref.26), the
Author addresses first commercially available stimulators were applied
to the treatment of pain by spinal cord stimulation
1
Department of Neurosurgery, Hannover Medical School, Hannover, Germany.
2
Department of Neurosurgery, Sunnybrook Health Sciences Centre, Toronto, ON, Canada. (SCS). Owing to the work of Mazars27 and Hosobushi28,
3
Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK. DBS targeting of the sensory thalamus soon followed,
4
Joint Department of Medical Imaging, University of Toronto, Toronto, ON, Canada. building on technology from the SCS hardware. DBS
5
Medical Research Council Brain Network Dynamics Unit, University of Oxford, Oxford, UK. for pain became widespread in Europe but was never
6
Department of Neurosurgery, Yonsei University College of Medicine, Seoul, South Korea. FDA-​approved for clinical use in the USA. The implant-
7
Department of Biomedical Engineering, Duke University, Durham, NC, USA. able hardware consisted of a DBS electrode and an
8
Department of Clinical Neuroscience, University of Umea, Umea, Sweden. extension cable, powered by a radiofrequency receiver
9
Department of Neurology, Movement Disorders and Neuromodulation Section, Charité and an external transmitter driven by a 9-​V battery and
Medicine University of Berlin, Berlin, Germany. carried by the patient. The 1970s also saw the intro-
10
Department of Neurosurgery, Zucker School of Medicine at Hofstra/Northwell,
duction of DBS in movement disorders to replace or
New York, NY, USA.
11
Department of Neurosurgery, Rutgers New Jersey Medical School, Newark, NJ, USA. complement the thalamotomies that were widely per-
12
Department of Neurosurgery, Stanford University, Stanford, CA, USA. formed at the time as well as, more rarely, in psychiat-
13
Department of Neurology, University Hospital of Würzburg, Würzburg, Germany. ric applications29. A rudimentary DBS design allowing
14
Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, closed-​loop adaptive stimulation was described as early
ON, Canada. as 1980 in patients with multiple sclerosis tremor30,31.

www.nature.com/nrneurol
Reviews

Lawrence Pool
performs first Benabid et al. DBS device Availability of
chronic DBS First fully report successful capable of DBS system
intervention using implantable treatment of simultaneous with wireless
a silver electrode cardiac DBS is mainly tremor with DBS, Rechargeable stimulation recording
connected to an pacemaker used for the ushering in the DBS batteries and recording capability and
induction coil introduced treatment of pain modern era of DBS available introduced 3 T compatibility

1947 1948 1958 1963 1970s 1980s 1987 1999 2002 2009 2013 2015 2020

Spiegel and Delgado uses a ‘External’ DBS First fully Dual channel Quadripolar Closed-loop Directional
colleagues develop ‘stimoceiver’ to systems in use, internalized DBS IPG, allowing electrode responsive DBS electrode
their stereotactic wirelessly inhibit with handheld systems available. two electrodes commercially DBS system commercially
frame158, followed aggressive radiofrequency Lithium batteries to be powered available introduced to available
in 1949 by the behaviour in transmitter greatly extend by one battery treat epilepsy
arc-based Leksell a bull159 implant life
frame

1.27 mm

0.5 mm

1.5 mm

Fig. 1 | Timeline of technology development for DBS. DBS, deep brain stimulation; IPG, implantable pulse generator.

In this system, activation of the deltoid muscles by the and Aleva Neurotherapeutics, entered the DBS market
Quadripolar electrodes
patient triggered the stimulation of the thalamic and with technical innovations such as segmented leads,
Deep brain stimulation (DBS)
electrodes configured with four subthalamic areas, which stopped the tremor. directional stimulation, longer battery duration, more
equally spaced contacts — the The modern era of DBS arrived in 1987, when flexibility in deciding stimulation parameters and
most commonly used DBS a group from Grenoble published their experience remote internet-​based programming. The most recent
electrode configuration. with DBS for essential tremor (ET) and PD tremor32. developments include current directionality with elec-
Radiofrequency coupled
Although quadripolar electrodes were already available in trode segmentation, increased parameter space for pro-
coils the 1970s, the electrodes initially used in Grenoble had gramming (in particular, shorter pulse widths (10 μs)
Early deep brain stimulation only one contact at the tip and chronic stimulation was and frequencies of up to 10,000 Hz) and advances in
systems powered the delivery achieved through radiofrequency coupled coils. The first MRI compatibility and neural recording capabilities.
of stimulation using an
implantable pulse generator (IPG) for DBS, manufactured
implanted radiofrequency
receiving coil. These systems by Medtronic, had a maximal frequency of 130 Hz — Innovation in electrode and IPG design
evolved and were replaced by the value used in most current DBS applications — and Electrode design. The basic principle of DBS is to use
the modern-​day battery- accommodated only unilateral stimulation. In 1999, the a small electrode to deliver electrical impulses to focal
coupled pulse generators. first dual-​channel IPG was launched in Europe. This brain regions. The crucial characteristics of an electrode
Implantable pulse generator
device delivered a current with a frequency of up to include biocompatibility, inertness, durability, stability
(IPG). A battery, typically 250 Hz and, with the increasing adoption of bilateral over time, surgical feasibility, good conductivity, electri-
implanted below the clavicle DBS globally, particularly for subthalamic nucleus cal properties, tractability, appropriate current delivery
and connected via subcutan­ (STN) stimulation in PD, it became the most used IPG and spatial configuration. Additional considerations
eous extension cables to intra­
worldwide. include MRI compatibility and the potential for sensing.
cranial electrodes. The IPG
generates and transmits elec- The next generation of DBS hardware was the Activa DBS electrodes consist of platinum–iridium wires
trical impulses at a specified series, namely Activa PC and Activa RC. These devices and nickel alloy connectors encased in a polyurethane
frequency, amplitude and expanded the parameter space that clinicians could use sheath. Platinum–iridium is chosen because of its mini-
pulse width. to programme DBS devices. Unlike previous IPGs, the mal toxicity and excellent conduction properties. Several
Parameter space
Activa IPG could be programmed to deliver either a con- electrode configurations are currently available (Fig. 2).
The available combinations of stant voltage or a constant current. Additional features The standard electrode configuration is quadripolar,
voltage, current, pulse width, included the possibility to deliver different stimulation with four stimulating electrode contacts at the tip of the
contact selection, current programmes in an interleaving pattern. Some of the probe, which is 1.27 mm in diameter. Each cylindrical
shape and stimulation pattern
newer IPGs were noted to have a shorter battery life than contact is 1.5 mm in length and contacts are spaced
when programming a deep
brain stimulation device. the earlier models33,34 and the relatively large size of the 0.5 mm or 1.5 mm apart. Such electrode configurations
implant and the need for replacement owing to battery allow the electric field to be shaped along the z axis of the
Segmented leads expiration every 3–4 years led to the introduction of lead through the programming of various combinations
Deep brain stimulation rechargeable IPGs. of anodes or cathodes.
electrodes with multiple
different contacts through
Starting around a decade ago, new companies, Since 2015, the availability of directional electrodes
which current can be including St. Jude Medical (subsequently acquired by has allowed more versatile shaping of the electric field,
transmitted. Abbott), Boston Scientific, SceneRay, PINS, Neuropace thereby improving the therapeutic window by enhancing

Nature Reviews | Neurology


Reviews

a Common DBS electrode configurations amplitudes lead to loss of directionality and the ability to
shape the stimulation field other than in the longitudinal
direction. Improvements in programming algorithms,
including a shift from trial and error to automated pro-
gramming, will be important to maximize the benefits
of new electrode designs.
Currently available commercial DBS systems are
manufactured in a labour-​intensive and cost-​intensive
process in which the electrodes are assembled manually.
Modern production techniques, such as film printing,
might increase the flexibility of electrode design and
allow further miniaturization but also raise concerns
about the long-​term performance and safety of novel
b Types of stimulation materials. The stability of impedance might be improved
by techniques such as nanocoating36,37.
Unipolar Bipolar Interleaving Multiple level Directional

Rapidly Biocompatibility. After implantation, an interface


alternating between the electrode and brain tissue develops, which
changes over time. In the chronic state, glial encapsula-
tion of the electrode, protein adsorption on electrode
sites and the characteristics of the ionic environment
at the electrode–electrolyte interface dictate the elec-
trical characteristics of the electrode–tissue interface38.
A gene­ral problem with the chronic implantation of
electro­des into the brain is an inflammatory foreign
body reaction, which needs to be minimized if stable
therapeutic responses are to be achieved with commer­
Fig. 2 | DBS electrode configurations. a | Common electrode configurations for deep cial deep brain electrodes 39. Studies of chronically
brain stimulation (DBS). Dark grey regions illustrate electrode contacts, which can be implan­ted leads have demonstrated a multinucleate giant
activated to deliver current. Electrode designs vary with regard to the spacing between
cell-type reaction irrespective of the duration of implan-
contacts as well as the number and shape of contacts. Greater contact spacing expands
the range of neural targets, whereas smaller contact spacing facilitates more precise
tation, which might be a response to the polyurethane
stimulation control. b | Modes of stimulation, depending on the type of DBS system in component of the surface coating of the electrode38.
use. Unipolar stimulation refers to current being directed from the battery to the contact Although these reactions require further study, the
or vice versa. Bipolar stimulation indicates current flowing between electrode contacts, global experience to date suggests that long-​term DBS
with at least one functioning as an anode and one as a cathode. Interleaving stimulation is reasonably safe.
refers to the alternation of different stimulation settings. Multiple level stimulation Idiopathic delayed-​onset oedema surrounding the
enables multiple neural targets to be stimulated, provided that they lie along the leads, presumed to be a subacute foreign body reac-
electrode trajectory. With directional stimulation, current can be directed or ‘shaped’ tion to electrode implantation, has occasionally been
on the basis of local anatomy or clinical symptoms. reported. The aetiology, predisposing factors and prog-
nosis of peri-​lead oedema are still unknown but it seems
efficacy and reducing adverse effects35. Unlike conven- to be more frequent than was originally assumed, with a
tional DBS electrodes, which use a cylindrical config- substantial proportion of cases being detected on routine
uration, directional electrodes use radially segmented postoperative MRI scans40.
contacts that allow the stimulation field to be moved in
the horizontal plane or shaped using anodes and cath- IPGs and programming. Innovation in IPG technology
odes to steer the current in a particular direction (Fig. 2). is long overdue in the DBS field. Advances in the shaping
The electrode and its capabilities need to be considered and control of current delivery, novel waveforms and pat-
as a unit with the technical properties of the connected terns, optimization of programming, energy efficiency,
IPG. Theoretically, single-​source, current-​driven or and miniaturization are necessary to enhance clinical
voltage-​driven devices allow less flexibility in the pro- outcomes, patient safety and comfort. Innovations that
gramming of electric fields than systems offering mul- have been in use for some time in SCS are now being
Electrode contacts tiple independent sources connected to each electrode adapted to DBS. Examples include multiple independent
Non-​insulated regions near the contact. current control, which involves the pairing of individual
distal tip of an electrode from Despite providing enhanced capabilities owing to lead contacts with a dedicated current source, thereby
which electrical impulses are
an increased number of available contacts, directional allowing the precise customization of stimulation field
transmitted.
electrodes add complexity to surgical implantation and size and shape. In addition to field shape optimization,
Waveforms create challenges for programming. The limits to the neuromodulation for pain has benefited from the use of
The shapes of the electrical benefits and feasibility of increasing contact numbers novel waveforms such as BurstDR and 10-​kHz high fre-
impulses transmitted from a are illustrated by the Sapiens electrode. This electrode quency (HF-10) therapy41,42; these stimulation platforms
deep brain stimulation contact,
most often represented in 2D
has 64 contacts, making integration with the extension are likely to be explored in DBS in the near future.
as a function of voltage or cable surgically challenging and has never been used for The optimization of programming through stand-
current over time. chronic stimulation (Fig. 2). In addition, higher current ardization is one of a number of different strategies that

www.nature.com/nrneurol
Reviews

are under investigation in the field of neuromodulation. Acute hardware failure might not only result in the
Both BurstDR and HF-10 run on standard algorithms rebound of symptoms such as tremor or depression
and the sometimes cumbersome and lengthy ‘art of but might also lead to severe adverse events such as
programming’ is being replaced with the ease of auto- the neuroleptic-​like malignant syndrome in PD49. The
mation. This approach is likely to make its way to DBS newer IPGs allow better readouts of battery capacity to
programming, with the ultimate goal of developing determine the date for pre-​emptive replacement and the
closed feedback loops and artificial intelligence-​based use of error-​detecting servomechanisms could also be
programming optimization. To some degree, we are considered in the future.
already seeing automatic programming in the SCS
world. For example, a teachable IPG that is available Advances in stimulation methods
for SCS uses position data from a built-​in accelerom- After a long period of stasis in hardware development,
eter to toggle through and automatically select optimal over the past few years, we have witnessed an abundance
pre-​programmed parameters43. The increasingly com- of new stimulation methods, including the introduction of
plex implanted electrode systems will require artificial regulated current IPGs, novel stimulation waveform
intelligence-​based or computational model-​based pro- shapes and novel temporal patterns of stimulation. These
gramming to achieve optimal results. A commercially technological improvements have led to a re-​evaluation
available system from Boston Scientific allows clinicians of stimulation algorithms and consideration of new
to designate a desired volume of tissue activated (VTA), paradigms of stimulation treatment, many of which are
after which the programming software will determine available but not yet sufficiently tested. Studies using
the scheme for contact activation. Although physician blinded comparisons of different stimulation paradigms
oversight of programming will always be required and are needed to assess their clinical utility50.
wireless DBS programming in the clinic is currently the
standard approach, remote monitoring and telemetry Controlled current versus controlled voltage. A consen-
applications as well as automated or self-​programming sus paper51 published in 2015 highlighted the paucity of
devices are likely to appear in the future. studies52,53 comparing clinical outcomes using regulated
Improvements in current delivery will need to be current DBS versus regulated voltage DBS but surmised
accompanied by changes to IPGs that make DBS more that, in the face of dynamic changes in load impedance,
palatable to prospective patients and physicians. These current DBS would be expected to produce more stable
changes include miniaturization and a reduction of the effects than voltage DBS54. As outlined above, the imped-
charging burden. Currently, the lightest IPG for SCS is ance of implanted DBS electrodes changes over time as a
29.1 g, whereas the typical DBS devices range from 40 g result of the inflammatory response and gliosis around
to 67 g. In addition, with renewed interest in recharge- the electrode55.
able DBS products, the charge time and capacity fade
become important. The currently available SCS IPGs Stimulation waveform shape. The stimulation wave-
charge from empty to full in 1 h and have >95% battery form — that is, the shape of the stimulation current
Volume of tissue activated capacity at 9 years but these properties have yet to be (or voltage) as a function of time — can influence the
(VTA). The estimated spatial
translated to DBS IPGs. Energy-​harvesting IPG tech- number and type of neural elements that are activated56
extent of the electric field
surrounding an activated deep nology has the potential to eliminate manual battery and waveforms or pulses can be repeated at various inter­
brain stimulation contact at a ­charging altogether44. pulse intervals to create a stimulation pattern (Fig. 3).
given stimulation parameter As IPGs become smaller, we should also expect the Comparisons of different stimulation waveforms and
setting. emergence of cranial or possibly even burr hole-​mounted patterns suggest that symmetric biphasic pulses produce
Energy-​harvesting
IPGs. Such technology would eliminate the risk of wire greater suppression of PD motor symptoms than do
Having the capability to passage and complications associated with wire breakage conventional asymmetric DBS waveforms with a long-​
capture energy from the but could also introduce new concerns such as cranial duration anodic recharge phase57, albeit at the expense
surrounding environment, IPG infections. of additional battery drain. Similarly, in patients with
including from thermal,
ET undergoing thalamic nucleus ventralis interme-
vibratory, electromagnetic and
acoustic sources. Patient safety concerns. Patient safety is a top priority in dius (Vim) DBS, symmetric biphasic pulses produced
neuromodulation. Full-​body MRI-​safe systems should a greater suppression of tremor than conventional
Biphasic pulses be an attainable goal for all manufacturers in the near asymmetric DBS waveforms58. At any given stimulation
Electrical impulses consisting future. Changes to the physical implant, including the intensity, symmetric biphasic pulses are likely to acti-
of both a positively and
a negatively charged
elimination of extension cables, would reduce concerns vate a larger number of neurons than asymmetric pulses,
component. During each regarding the heating of DBS systems during MRI scans. as both the cathodic and anodic phases of the stimulus
stimulus, a reversal between The prevention of infection of implantable devices is also waveform can contribute to net activation59.
cathodic and anodic essential and current infection rates in patients with Other factors that might improve the activation
stimulation occurs.
chronic DBS range from 5–10%45–47. We are already see- and entrainment of neurons include an appropriate
Cathodic and anodic ing the use of antibacterial envelopes to prevent cardiac choice of waveform polarity, reversal of the standard
During stimulation, an pacemaker infections. Indeed, a recent randomized con- pulse phase order, and a gap between the two phases of
electrode contact can function trolled study in people with cardiac implantable devices charge-​balanced biphasic pulses60. The waveform shape
as a cathode (or current sink) showed that the use of antibacterial envelopes signifi- can also influence the desynchronizing impact of DBS
or as an anode (source of
current) relative to the
cantly reduced infection rates48. The antibiotic coating techniques in which the pulse amplitude is modulated
implantable pulse generator or of neurostimulation systems could also prevent infection in a closed-​loop manner by linear or nonlinear delayed
to other electrode contacts. and the subsequent need for explantation. feedback61–63.

Nature Reviews | Neurology


Reviews

a b c d strong or weak synapses and synchrony75. In theoreti-


cal models and simulations, desynchronizing stimula-
tion reduces neuronal coincidence rates which, mediated
by spike timing-​dependent plasticity, might decrease
synaptic strength75,77,78. Coordinated reset stimula-
e f g h tion, which delivers brief high-​frequency pulse trains
through different stimulation contacts74, might cause
acute desynchronizing effects during stimulation as
well as unlearning of abnormal neuronal synchrony
and synaptic connectivity. Desynchronization was
Fig. 3 | Stimulation waveform shapes and temporal stimulation patterns. In deep shown to accumulate and persist after the cessation
brain stimulation (DBS), waveform shapes are repeated at interpulse intervals to create of coordinated reset stimulation75,77,78 and long-​lasting
a stimulation pattern. a | Conventional asymmetric biphasic DBS waveform with a effects were verified in both preclinical and clinical
short-​duration cathodic phase followed by an interphase delay and a long-​duration studies79–81.
anodic (recharge) phase. b | Symmetric biphasic DBS waveform with equal-​duration These emerging and promising results indicate the
cathodic and anodic phases. c | Symmetric biphasic DBS waveform with zero interphase potential of temporal patterns to achieve an enhanced
delay. d | Reversal of the standard pulse phase order of a symmetric biphasic DBS efficiency of stimulation and prolong symptom relief
waveform. e | Regular temporal pattern of stimulation with fixed interpulse intervals
through the control of plasticity.
(typically ~7.7 ms or ~130 Hz). f | Non-​regular temporal pattern of stimulation with
random interpulse intervals. g | Burst pattern of stimulation with several pulses at short
interpulse intervals followed by a long interpulse interval. h | Stimulation pattern for Adaptive DBS and closed-​loop systems
coordinated reset with bursts of stimulation distributed across four different electrode The possibility of regulating DBS over time according
contacts, with each row corresponding to the stimulation pattern delivered to each to one or more feedback signals has attracted consid-
electrode contact. erable interest in recent years. This approach, which we
term ‘adaptive DBS’, encompasses responsive, adaptive
and closed-​loop control modes82,83. The development
Similar to early studies of Vim DBS for tremor64, the of adaptive DBS is primarily motivated by the poten-
thresholds for STN DBS to produce both adverse effects tial for improved efficacy and reduced risk of adverse
and reductions in PD motor symptoms were higher with effects. A simple example is position-​adaptive SCS treat-
anodic stimulation than with cathodic stimulation65. ment for pain, whereby feedback from an accelerometer
However, at amplitudes just below the adverse effect automatically adjusts the stimulation voltage according
threshold, anodic stimulation produced a greater to positional changes in the electrode with respect to
suppression of symptoms than cathodic stimulation. its target43. However, in most applications, feedback
relates directly or indirectly to the dynamic state of the
Stimulation patterns. Increasing evidence suggests nervous system. Although a mechanistic relationship
that the temporal pattern of stimulation can influence between the feedback signal and neural dysfunction is
the clinical outcome after DBS66, particularly in PD67. not essential, causally relevant signals have the advan-
The selection of an optimal pattern creates a substantial tage that they can change before symptoms emerge,
design challenge: assessing the effects of a stimulation potentially allowing predictive rather than reactive
pattern on symptoms is difficult enough when the symptom management. To date, adaptive DBS has
responses to stimulation are relatively rapid and overt, been studied mainly in PD, tremor and epilepsy but is
for example, in the case of tremor in ET68 or bradykinesia increasingly being explored in other conditions such as
in PD69. However, this assessment becomes exceedingly Tourette syndrome82,84.
challenging in instances where the outcomes are slow Local field potentials (LFPs) recorded from con-
Spike timing-​dependent to develop and cannot readily be observed such as in tacts of implanted electrodes have revealed differences
plasticity dystonia, where improvement of tonic symptoms is often in power spectra in various disorders 85–91 (Table 1) .
Concept by which the timing of
seen only after a delay70, or in epilepsy, where changes in Adaptive DBS in PD has focused on LFP feedback in
presynaptic and postsynaptic
excitatory potentials affects seizure frequency might occur only after several months. the beta frequency range as a correlate of bradykinesia
the overall synaptic strength. In addition, the response to stimulation might not be sta- and rigidity87,88 and on gamma activity, picked up from
ble over time as has been observed in ET. Furthermore, a cortical strip electrode, as a marker of dyskinesia85
Neuronal coincidence rates the range of possible temporal patterns is very large (Fig. 4). Beta activity recordings can be smoothed over
The incidence of temporally
overlapping presynaptic and
and an empirical approach to identifying the most clin- many seconds88 or processed to retain rapid fluctua-
postsynaptic excitatory ically effective pattern may not be feasible. One alter- tions in the signal87. The heavily smoothed signal pre-
potentials. native is to employ model-​based optimization of the dominantly tracks the dynamics related to drug therapy
temporal pattern71; however, this approach requires and captures motor on–off state changes. The use of
Power spectra
a high-​f idelity model of the relationship between this form of feedback to drive DBS can reduce power
In the context of local field
potentials, it refers to the the pattern of stimulation and changes in a particular requirements by ~50% and has been shown to reduce
strength or intensity of symptom67,72. on-​state dyskinesias92.
the electric field based Stimulation techniques have been computationally Alternatively, beta feedback can be processed to
on frequency, commonly designed to counteract the abnormal synchronization capture the bursting nature of spontaneous beta activ-
categorized as delta (1–3 Hz),
theta (4–8 Hz), alpha (4–9 Hz),
of neuronal activity73–75. In the presence of spike timing-​ ity. This strategy is based on growing evidence that
beta (15–30 Hz) and gamma dependent plasticity 76, neuronal populations display these bursts, particularly when prolonged and elevated
(>30 Hz). complex dynamics and can stably reside in states with in amplitude, mediate bradykinesia and rigidity89,93. In this

www.nature.com/nrneurol
Reviews

application, the detection of longer-​duration bursts acceptably safe101. In 2019, the RNS system was com-
triggers or increases stimulation to terminate these bined with stimulation of the anterior nucleus of the
bursts. This approach also achieves a 50% reduction thalamus with the aim of extending the use of the sys-
in power requirements and reduces adverse effects on tem to refractory multifocal or generalized epilepsy102.
speech in comparison with conventional continuous Indeed, an adaptive approach might help ameliorate the
stimulation87,94,95. It has also achieved better control frequent arousals from sleep that are experienced with
of bradykinesia and rigidity than adaptive DBS based conventional stimulation of this target103. Another devel-
on smoothed beta feedback88 but the control of dyski- opment has been the tachycardia-​based seizure detec-
nesia has not been objectively confirmed. An alterna- tion algorithm implemented in the AspireSR system,
tive approach to control dyskinesia is to detect rapid which allows automatic top-​up vagal nerve stimulation
changes in finely tuned gamma activity at the cortical during seizures104,105.
level, where its amplitude is larger and more easily dis- Despite excitement about the prospect of dynamic
tinguished from stimulation artefacts than in other areas interfacing with the brain to control symptoms,
of the brain85. position-​adaptive SCS for pain and RNS for epilepsy are
Adaptive DBS for tremor is being explored using the only approaches that have made substantial inroads
LFP activity recorded from the STN, cortical surface or into clinical practice so far. This situation is likely to
thalamus or using one or more electromyographic or kin- change in the near future as flexible bidirectional devices
ematic sensors on the body for feedback86,96–98. Features for chronic implantation become more widely available,
extracted from LFPs have been used to decode the onset allowing chronic trials of novel adaptive approaches with
of tremor and, in the case of ET, tremor-​triggering rescue in the form of standard DBS as necessary as well
voluntary movement86,96,99. as long-​term recording of neurophysiological signals for
Though exciting, the application of adaptive DBS in machine learning of the precise correlations between
movement disorders is at an early stage and the efficacy neural activity and symptom severity across a variety of
and adverse effect profiles remain to be determined in diseases. Another area of increasing interest is implan-
patients with chronic implants. In addition, it remains tation and recording from more than one brain site in
to be seen whether feedback control can be too selec- individual patients106, thereby ameliorating the effects
tive in some patients, necessitating the use of two or of stimulation artefacts and enabling network-​related
more control loops for different symptoms. Such an rather than single-​site feedback control85,107. Feedback
approach might prove necessary in the few patients control is also likely to expand to include multiple
with PD who develop breakthrough tremor during signals related to the neural and physical state, sup-
adaptive beta-​based DBS for bradykinesia and rigidity100. ported by more sophisticated control algorithms.
Opportunities to develop more sophisticated adaptive These advances could be facilitated by the development
DBS approaches based on computational modelling are of local and distributed cloud computing systems108
also arising. and firmware upgrades to allow ‘future-​proofing’
In the epilepsy field, considerable experience has been of implants109.
gained with the NeuroPace responsive neurostimulation
(RNS) system, which includes a cranium-​implanted DBS imaging and stimulation modelling
pulse generator connected to one or two recording and Over the past decade, advances in neuroimaging have
stimulating depth and/or cortical strip electrodes placed improved DBS target visualization and lead localization
at previously identified seizure foci. The pulse genera- (Fig. 5). These developments have informed both surgical
tor provides short-​term stimulation in response to the targeting and postoperative DBS programming. Novel
detection of abnormal electrocorticographic activ­ imaging methods have also led to a better understanding
ity that precedes a possible clinical seizure. This stim- of the mechanisms of action of DBS.
ulation approach is reported to reduce the frequency Some DBS targets are poorly visualized on the brain
of partial-​onset seizures and is well tolerated and MRI scans that are routinely acquired for surgical

Table 1 | Neural biomarkers of local field potentials


Neural biomarker recording site Disease Main uses refs
Gamma (~70 Hz) activity Cortex Parkinson Feedback signal related to dyskinesia for 85

disease adaptive DBS


Beta (~20 Hz) activity Cortex Essential Triggering stimulation in response to 86

tremor movement in adaptive DBS


Beta (~20 Hz) activity Subthalamic nucleus or Parkinson Feedback signal related to bradykinesia– 87,88,89

globus pallidus internus disease rigidity for adaptive DBS; localizing signal
for contact selection in programming
Theta (~7 Hz) activity Subthalamic nucleus or Dystonia Feedback signal related to muscle spasms 90

globus pallidus internus for adaptive DBS


Event-​related Evoked potentials from Parkinson Localizing signal for contact selection in 91

resonance activity subthalamic nucleus disease programming


(~260 Hz) stimulation
DBS, deep brain stimulation.

Nature Reviews | Neurology


Reviews

a Beta amplitude High beta correlates with because clinicians must decide how to steer the current
DBS slowness and stiffness during programming119. Several software tools have been
ECOG electrode
strip introduced for this purpose113,120–123, and algorithms
that reconstruct electrode localization from CT and
DBS MRI scans have been developed121,124–126. Furthermore,
STN
algorithms were created and validated to reconstruct
Time
the orientation of segmented leads127. Electrode locali-
b Gamma amplitude zation is particularly important as evidence suggests that
High gamma co-occurs the segmented contacts of directional DBS leads often
with dyskinesias
show large deviations from their intended implantation
orientation119.
Retrospective group-​level analysis of precise elec-
DBS
trode locations in multiple patients provides an oppor-
tunity to further our understanding of DBS mechanisms
Time of action128. Group-​level analyses have been made pos-
Fig. 4 | Adaptive DBS in Parkinson disease. a | Stimulating and recording from the sible by several recent advances in neuroimaging: first,
deep brain stimulation (DBS) electrode. When the patient is in the off-​levodopa phase, the precise co-​registration of patients’ brains onto an
which is characterized by slow movements and muscle stiffness, the local field potential average brain template (for example, by non-​linear
activity at the electrode contacts contains prominent oscillations at ~20 Hz (beta activ­ normalization to the Montreal Neurological Institute
ity). The amplitude of these oscillations varies over time and high-​frequency DBS is brain template)129; second, accurate DBS electrode
delivered whenever an amplitude threshold is crossed. An alternative approach is to localization;113 and third, estimation of the VTA130–132.
deliver DBS with an intensity that is proportional to the beta amplitude. b | Stimulating Large sample sizes can be used in group-​level analyses
DBS electrode and recording from an electrocorticographic (ECOG) electrode strip to estimate robust ‘sweet spots’ or optimal connectiv-
overlying the motor cortex. When the patient is on levodopa and dyskinetic, the ECOG ity profiles that could be useful to predict outcomes in
activity picked up by the strip electrode contains prominent oscillations at ~70 Hz
future patients. Probabilistic maps of clinical outcomes
(gamma activity). The amplitude of these oscillations is monitored and DBS is stopped
or reduced when an amplitude threshold is crossed. STN, subthalamic nucleus. and efficacious networks can be computed with clini-
cally weighted contact locations or VTAs to pinpoint
the most effective neuroanatomical substrates across
planning. For instance, although the STN is visible a large cohort of patients. Of note, the VTA is a visual
on T2-​weighted sequences, its ventral border towards approximation that is inferred from a model-​based the-
the substantia nigra is often difficult to delineate. The oretical concept and its validity depends on the model
inter­nal medullary lamina separating the globus pal- used. In addition, estimation of the VTA ignores local
lidus internus and externus is not visible on routine impedance changes and intrinsic dynamics of neuronal
T1-​weighted sequences. To address these limitations, populations.
higher field strengths and novel sequences have been Such methods have enabled direct relationships to
developed to improve the visualization of DBS targets110. be established between electrode placement (and stim-
For example, quantitative susceptibility mapping applied ulation location) and clinical improvement in PD133–137,
to gradient-​echo sequences is highly sensitive to iron dystonia138–140, ET141,142 and OCD143 (Fig. 5). This work has
and provides striking improvements in STN visual- led to the emergence of sweet spots — that is, optimal
ization111 (Fig. 5). Similarly, the fast grey matter acqui- surgical and stimulation targets — that have direct and
sition T1 inversion recovery sequence was introduced statistically significant relationships with clinical out-
to enhance the visualization of subcortical structures112 comes. For instance, studies by several different groups
and is specifically optimized to visualize the globus worldwide have converged on an optimal target for the
pallidus internus and substructures of the thalamus113. treatment of PD144.
Diffusion-​weighted imaging is also gathering interest as In addition to defining brain areas implicated in
a targeting tool focusing on white matter tracts, particu- clinical outcomes, the inclusion of information about
larly for ET treatment114. Ultra-​high-​field (UHF) MRI at functional or structural connectivity has led to the con-
7 T also holds promise for improving target visualization. cept of network-​based or tract-​based targets137,145,146.
For example, the intrathalamic nuclei can be seen with For example, modulation of targets structurally con-
UHF MRI115,116, which is a notable advantage when plan- nected to the supplementary motor area was shown
ning DBS surgery for tremor (Fig. 5). STN borders can to be associated with clinical improvement in PD137.
also be better appreciated on UHF MRI than on conven- The connection profiles could be further differentiated
tional MRI117,118. A fundamental limitation of UHF MRI according to symptoms: connectivity from the active
is an increased susceptibility to distortion artefacts, espe- contact within the STN to the supplementary motor
cially in the centre of the brain, which requires c­ areful area explained bradykinesia and rigidity improve-
distortion correction. ment, whereas the alleviation of tremor was associated
Electrode localization is paramount to confirm ade- with connectivity to the primary motor cortex134. Differ­
quate targeting and to define the neural substrates that ential connectivity was also associated with discrete
are responsible for clinical outcomes. The reconstruction tremor improvement in ET141. Structural connectivity
of precise electrode placements relative to surrounding between the active contact within the Vim and the hand
anatomical structures is particularly important with the regions of the primary motor cortex (and the cerebel-
rise in popularity of segmented and directional leads lum) was associated with improvement in hand tremor,

www.nature.com/nrneurol
Reviews

whereas connectivity-​mediated modulation of head connecting to the STN was predictive of efficacy of the
regions led to improvement in head tremor141. anterior limb of the internal capsule or STN DBS in two
Connectivity might play an even greater role in psy- OCD patient cohorts149. This last example demonstrates
chiatric surgery, for which the targets are not yet univer- how different DBS targets used to treat the same disease
sally agreed. The establishment of efficacious connectivity can potentially modulate a common tract or network,
profiles could aid the refinement and adjustment of sur- thereby alleviating a similar set of symptoms.
gical targets143,147,148. A 2019 report demonstrated direct As we have discussed in this section, retrospective
relationships between clinical improvement and connec- studies exploring the relationships between stimula-
tivity profiles of DBS electrodes in patients with OCD143. tion location and clinical outcomes can yield useful
The authors identified a subregion within the anterior information regarding the mechanisms of action of
limb of the internal capsule that was associated with DBS. However, prospective data acquisition in patients
good clinical improvement. Importantly, a second study undergoing DBS (for example, functional MRI with DBS
showed that the proximity of the electrode to a white turned on) could provide a more direct in vivo insight
matter bundle coursing through this subregion and into the neural changes induced by DBS. To date, the

a b c
CeM
MTT VA
IC VLA
VLP
MD
CM
VPL
PuM
PuL

DBS system QSM 7 T imaging


d e
Functional connectivity

Structural connectivity

Lead localization and VTA Connectomic mapping

Fig. 5 | DBS neuroimaging. a | Postoperative neck and chest X-​rays showing an implanted deep brain stimulation (DBS)
system with electrodes and extension wires (left image) and the implantable pulse generator implanted over the chest
area (right image) b | Novel MRI visualization techniques, including quantitative susceptibility mapping (QSM) and fast
grey matter acquisition T1 inversion recovery, have improved the visualization of subcortical structures. QSM coronal
slice shows the subthalamic nucleus (outlined) — the most commonly targeted structure in Parkinson disease. c | Ultra-
high-​field preoperative MRI is increasingly used in surgical planning and research. T1-​weighted axial slice intrathalamic
nuclei (labelled in the right-​hand image). d | On the basis of MRI metallic artefacts associated with DBS electrodes
(shown on T2-​weighted coronal image and axial image (arrowheads) on the left), the electrodes can be localized and
reconstructed in 3D using specialized software. CT scans can also be used for electrode localization. DBS settings,
including active contact, voltage, pulse width and impedance, can be used to estimate the electric field (white arrows,
right image) surrounding the DBS electrodes. Heuristic assumptions or axonal cable models can be used to estimate the
volume of tissue activated (VTA, red, right image). e | The VTA can be used in connectivity analyses informed by metrics
such as resting-​state functional MRI (top left) and diffusion-​weighted, imaging-​based tractography (bottom left) to
determine the effects of DBS on distributed brain regions. CeM, central medial nucleus; CM, centromedian nucleus;
IC, internal capsule; MD, mediodorsal nucleus; MTT, mammillothalamic tract; PuM, medial pulvinar nucleus; PuL, lateral
pulvinar nucleus; VA, ventral anterior nucleus; VLA, ventral lateral anterior nucleus; VLP, ventral lateral posterior nucleus;
VPL, ventral posterolateral nucleus. Part a is adapted with permission from ref.152, Boutet, A. et al. Radiology (2019) 293,
174–183, Radiological Society of North America. Part b is adapted with permission from ref.110, Liu, T. et al. Radiology
(2013) 269, 216–223, Radiological Society of North America. Part c is adapted with permission from ref.115, Elsevier. Part e
is adapted with permission from ref.137, Wiley.

Nature Reviews | Neurology


Reviews

prospective acquisition of functional neuroimaging and programming devices and the incorporation of
data in patients with fully internalized and active DBS commercial platforms into the systems, create a real
has been limited by the inherent risks of exposure of risk of failure of device security14,153. In his considera-
active implantable medical devices to the magnetic tions on ‘posthuman capitalism’, contemporary phi-
fields in MRI scanners150. Generally, these studies were losopher Slavoj Zizek outlined and summarized some
hampered by MRI acquisition restrictions (for exam- of the potential dangers of misusing various types of
ple, magnet strengths of no more than 1.5 T) owing to brain–machine interfaces154. In particular, the con-
safety guidelines. However, recent investigations have cept of directly coupling neural circuits with software
shown safe and feasible acquisition of functional MRI on digital devices could bear unforeseeable risks. The
using 3 T (with a body transmit coil) in large cohorts hacking of digital technology and programmes has
of patients with DBS systems50,151,152, enabling a broader entered new dimensions over the past few years and, in
range of functional neuroimaging data acquisition and the age of the ‘Internet of things’, technology dysfunc-
opening the door to a new field of neuromodulation tion caused by “distributed denial of service” attacks
research. has become a reality155. As suggested in a 2019 article in
The Economist, “a connected world will be a playground
DBS in a connected world for hackers”156.
The advantages offered by integrating electronics with Hacking has reached the level of global social and
the human nervous system are substantial. The ability to political manipulation and data from chronic neural
collect extensive datasets from large numbers of patients recordings made available via cloud technology, in par-
undergoing DBS is likely to have a positive impact on ticular from limbic and cognitive-​associative circuit-
both patient care and the development of neuromod- ries, could potentially be misused not only to ‘optimize’
ulation platforms but also raises concerns over safety, emotional and cognitive states but also to establish new
privacy and security. forms of social control. Although such ‘neurosecu-
Improvements in technology as well as the introduc- rity’ threats are still mostly theoretical, we believe that
tion of wireless connectivity to interact with monitoring a discussion of these issues should be initiated before
they become apparent. Brainjacking, a term coined by
Box 1 | Anticipated advances in deep brain stimulation technology Pycroft, is a potentially serious threat that warrants
Improvements in electrode and IPg design early discussion before any real-​world harms occur14.
• Miniaturization and cranialization As a result of the paucity of work specifically address-
• Large-​scale production and modernized production techniques to reduce cost ing neurosecurity and brainjacking, several areas of
ethical consideration and consultation might prove
• Multiple power sources within implantable pulse generators (IPGs) to enable multiple
independent current control
fruitful together with greater investment in security
measures.
• Improved battery life, recharging capacity or energy harvesting
Various ways of attacking neurostimulation sys­
Increased safety tems can be envisaged. Simply draining the battery or
• Improved MRI compatibility with ≥3 T systems switching off the device could cause rebound symptoms
• Antibiotic impregnation to reduce infection or tissue damage or attacks might be targeted to spec­
• Protection from hacking, including ‘brainjacking’ ific indications, leading to motor impairment, increased
pain, altered impulse control or unpleasant emo­tions.
optimized stimulation
The philosophical implications of exerting control over
• IPGs with multiple independent power sources to enable multiple independent
current controls
another human being in this manner could be profound
and deserve further analysis. The legal and economic
• Increased control over waveform shape (for example, symmetric biphasic pulses for
tremor)
implications may also be substantial given the expected
proliferation of neurotechnology in the coming years.
• Use of varying interpulse intervals or coordinated reset (brief high-​frequency pulse
trains from different contacts)
Ongoing discussion and study of these threats among
clinicians and industry is important to minimize the
Adaptive or closed-​loop designs risks. Clinicians should educate themselves about
• Stimulation modulated by body position (gyroscopes) or motion (accelerometer) the basics of information security and be mindful of the
• Response to local field potential power spectra (for example, beta for rigidity or risks of brainjacking when evaluating faulty implants
gamma for dyskinesia) or seizure activity or caring for high-​profile patients. Hospital staff should
• Electromyographic recording for motor feedback also be aware of social engineering techniques used by
• Integration of multiple feedback and stimulation sites attackers to gain privileged information and should
• Use of artificial intelligence techniques to fine-​tune programming have at least a basic understanding of how to minimize
neurosecurity risks. Patients should have some degree
Neuroimaging advances to improve targeting
of awareness of particularly risky behaviours to avoid,
• Enhanced anatomical resolution through specialized sequences (for example,
although any discussion of this topic should avoid undue
quantitative susceptibility mapping) or ultra-​high-​field (7 T) MRI
alarm and emphasize, at least at the present time, the
• Improved automatic electrode reconstruction and segmentation with
extremely low probability of an individual patient being
image-​processing software
targeted by an electronic attack.
• Identification of ‘sweet spots’ from large retrospective imaging studies
The history of both information security and med-
• Enhanced deep brain stimulation programming through prospective functional icine has amply demonstrated that prevention is better
imaging (for example, functional MRI) to identify optimal ‘neural signatures’
than cure and the best approach is to apply lessons to

www.nature.com/nrneurol
Reviews

a Current DBS systems b Future DBS systems neurosecurity while the situation remains tractable; the
Electrode Implantable pulse necessary developments include codes of best practice
• Single or bilateral electrodes generator for neurosecurity tailored to both the devices and the
• Continuous stimulation • Smaller, cranialized indications157. Close cooperation between stakeholders,
• Connected to Wi-Fi
• Longer battery life
such as clinicians, patients and device manufacturers, is
• Rechargeable or paramount but should allow flexibility to enable device
Extension cables
energy-harvesting development.

Electrodes Conclusions and future directions


• Multiple leads, Our understanding of the brain network circuit mal-
capable of sensing
and stimulating functions that lead to clinical manifestations of neurol­
• Closed-loop/adaptive ogical and psychiatric diseases is increasing, with these
insights informing novel DBS hardware design and
stimulation methods. We envisage a future in which
Implantable pulse neuromodulation will be safer, less invasive, and more
generator
• Lithium battery in accurate and efficacious, and will be applied to a greater
titanium housing No extension cables proportion of patients for whom other forms of treat-
• Adjusted with physician ment have proved insufficient. In particular, we antic-
programmer
• 3–5-year battery life ipate advances in electrode design, IPG capabilities,
and programming and stimulation methods (Box 1;
Fig. 6). Sophisticated imaging techniques will improve
the identification of brain targets, validate target
engagement and confirm the attainment of the desired
physiological circuit effect of stimulation. However,
as with other powerful and life-​changing technolo-
gies, ethical, privacy and security safeguards are of the
utmost importance and must be considered in paral-
lel with technological progress to avoid unintended
consequences.
Fig. 6 | Future visions for DBS. a | A typical currently available deep brain stimulation
(DBS) configuration b | A predicted future DBS configuration. Published online xx xx xxxx

1. Lozano, A. M. et al. Deep brain stimulation: current persons and systems. Camb. Q. Healthc. Ethics 25, disorders. Parkinsonism Relat. Disord. 16, 429–433
challenges and future directions. Nat. Rev. Neurol. 15, 623–633 (2016). (2010).
148–160 (2019). 14. Pycroft, L. et al. Brainjacking: implant security issues 26. Melzack, R. & Wall, P. D. Pain mechanisms: a new
2. Fasano, A., Aquino, C. C., Krauss, J. K., Honey, C. R. in invasive neuromodulation. World Neurosurg. 92, theory. Science 150, 971–979 (1965).
& Bloem, B. R. Axial disability and deep brain 454–462 (2016). 27. Mazars, G., Mérienne, L. & Cioloca, C. Use of thalamic
stimulation in patients with Parkinson disease. 15. Coffey, R. J. Deep brain stimulation devices: a brief stimulators in the treatment of various types of pain
Nat. Rev. Neurol. 11, 98–110 (2015). technical history and review. Artif. Organs 33, [French]. Ann. Med. Interne 126, 869–871 (1975).
3. Moro, E. et al. Efficacy of pallidal stimulation in 208–220 (2009). 28. Hosobuchi, Y., Adams, J. E. & Rutkin, B. Chronic
isolated dystonia: a systematic review and meta-​ 16. Pool, J. L. Psychosurgery in older people. J. Am. thalamic stimulation for the control of facial
analysis. Eur. J. Neurol. 24, 552–560 (2017). Geriatr. Soc. 2, 456–466 (1954). anesthesia dolorosa. Arch. Neurol. 29, 158–161
4. Limousin, P. & Foltynie, T. Long-​term outcomes of 17. Delgado, J. M. et al. Intracerebral radio stimulation (1973).
deep brain stimulation in Parkinson disease. Nat. Rev. and recording in completely free patients. J. Nerv. 29. Hariz, M. I., Blomstedt, P. & Zrinzo, L. Deep brain
Neurol. 15, 234–242 (2019). Ment. Dis. 147, 329–340 (1968). stimulation between 1947 and 1987: the untold
5. Fontaine, D., Vandersteen, C., Magis, D. & 18. Delgado, J. M., Obrador, S. & Martin-​Rodriguez, J. G. story. Neurosurg. Focus. 29, E1 (2010).
Lanteri-Minet, M. Neuromodulation in cluster in Surgical Approaches in Psychiatry (eds Laitinen, L. 30. Brice, J. & McLellan, L. Suppression of intention
headache. Adv. Tech. Stand. Neurosurg. 42, 3–21 & Livingston, K. E.) 215–223 (Medical and Technical tremor by contingent deep-​brain stimulation. Lancet
(2015). Publishing, 1973). 1, 1221–1222 (1980).
6. Pereira, E. A. & Aziz, T. Z. Neuropathic pain and deep 19. Heath, R. G. Electrical self-​stimulation of the 31. Blomstedt, P. & Hariz, M. Closed loop stimulation for
brain stimulation. Neurotherapeutics 11, 496–507 brain in man. Am. J. Psychiatry 120, 571–577 tremor was invented in 1980. Brain Stimul. 12,
(2014). (1963). 1072–1073 (2019).
7. Lee, D. J., Lozano, C. S., Dallapiazza, R. F. 20. Heath, R. G. Modulation of emotion with a brain 32. Benabid, A. L., Pollak, P., Louveau, A., Henry, S.
& Lozano, A. M. Current and future directions of pacemamer. Treatment for intractable psychiatric & de Rougemont, J. Combined (thalamotomy and
deep brain stimulation for neurological and psychiatric illness. J. Nerv. Ment. Dis. 165, 300–317 (1977). stimulation) stereotactic surgery of the VIM thalamic
disorders. J. Neurosurg. 131, 333–342 (2019). 21. Bickford, R. G., Petersen, M. C., Dodge, H. W. Jr. nucleus for bilateral Parkinson disease. Appl.
8. Mallet, L. et al. Subthalamic nucleus stimulation in & Sem-​Jacobsen, C. W. Observations on depth Neurophysiol. 50, 344–346 (1987).
severe obsessive-​compulsive disorder. N. Engl. J. Med. stimulation of the human brain through implanted 33. Kiss, Z. H. T. & Hariz, M. “New and improved” DBS
359, 2121–2134 (2008). electrographic leads. Proc. Staff. Meet. Mayo Clin. batteries? Brain Stimul. 12, 833–834 (2019).
9. Harmsen, I. E. et al. Clinical trials for deep brain 28, 181–187 (1953). 34. Hariz, M. Battery obsolescence, industry profit
stimulation: current state of affairs. Brain Stimul. 13, 22. Sem-​Jacobsen, C. W. Depth-​electrographic and deep brain stimulation. Acta Neurochir. 161,
378–385 (2020). observations related to Parkinson’s disease. Recording 2047–2048 (2019).
10. Deeb, W. et al. Proceedings of the Fourth Annual Deep and electrical stimulation in the area around the third 35. Steigerwald, F., Muller, L., Johannes, S., Matthies, C.
Brain Stimulation Think Tank: a review of emerging ventricle. J. Neurosurg. 24 (Suppl. 1), 388–402 & Volkmann, J. Directional deep brain stimulation
issues and technologies. Front. Integr. Neurosci. 10, (1966). of the subthalamic nucleus: A pilot study using a
38 (2016). 23. Bechtereva, N. P., Bondartchuk, A. N., Smirnov, V. M., novel neurostimulation device. Mov. Disord. 31,
11. Cagnan, H., Denison, T., McIntyre, C. & Brown, P. Meliutcheva, L. A. & Shandurina, A. N. Method of 1240–1243 (2016).
Emerging technologies for improved deep brain electrostimulation of the deep brain structures in 36. Angelov, S. D. et al. Electrophoretic deposition of
stimulation. Nat. Biotechnol. 37, 1024–1033 (2019). treatment of some chronic diseases. Confin. Neurol. ligand-​free platinum nanoparticles on neural
12. Ramirez-​Zamora, A. et al. Proceedings of the Sixth 37, 136–140 (1975). electrodes affects their impedance in vitro and
Deep Brain Stimulation Think Tank modulation of 24. Bechtereva, N. P., Kambarova, D. K., Smirnov, V. M. in vivo with no negative effect on reactive gliosis.
brain networks and application of advanced & Shandurina, A. N. in Neurosurgical Treatment in J. Nanobiotechnology 14, 3 (2016).
neuroimaging, neurophysiology, and optogenetics. Psychiatry, Pain, and Epilepsy (eds Sweet, W. H. et al.) 37. Koenen, S. et al. Optimizing in vitro impedance and
Front. Neurosci. 13, 936 (2019). 581–613 (Univ. Park Press, 1977). physico-​chemical properties of neural electrodes by
13. Kellmeyer, P. et al. The Effects of closed-​loop medical 25. Blomstedt, P. & Hariz, M. I. Deep brain stimulation electrophoretic deposition of Pt nanoparticles.
devices on the autonomy and accountability of for movement disorders before DBS for movement Chemphyschem 18, 1108–1117 (2017).

Nature Reviews | Neurology


Reviews

38. Kronenbuerger, M. et al. Brain alterations with deep delayed feedback for closed-​loop deep brain 87. Little, S. et al. Adaptive deep brain stimulation
brain stimulation: new insight from a neuropathological stimulation. PLoS One 12, e0173363 (2017). in advanced Parkinson disease. Ann. Neurol. 74,
case series. Mov. Disord. 30, 1125–1130 (2015). 62. Popovych, O. V., Lysyansky, B. & Tass, P. A. Closed-​loop 449–457 (2013).
39. Moss, J., Ryder, T., Aziz, T. Z., Graeber, M. B. deep brain stimulation by pulsatile delayed feedback 88. Arlotti, M. et al. Eight-​hours adaptive deep brain
& Bain, P. G. Electron microscopy of tissue adherent with increased gap between pulse phases. Sci. Rep. 7, stimulation in patients with Parkinson disease.
to explanted electrodes in dystonia and Parkinson’s 1033 (2017). Neurology 90, e971–e976 (2018).
disease. Brain 127, 2755–2763 (2004). 63. Popovych, O. V. & Tass, P. A. Multisite delayed 89. Tinkhauser, G. et al. Directional local field potentials:
40. Fenoy, A. J., Villarreal, S. J. & Schiess, M. C. Acute and feedback for electrical brain stimulation. Front. a tool to optimize deep brain stimulation. Mov. Disord.
subacute presentations of cerebral edema following Physiol. 9, 46 (2018). 33, 159–164 (2018).
deep brain stimulation lead implantation. Stereotact. 64. Benabid, A. L. et al. Chronic electrical stimulation of 90. Piña-​Fuentes, D. et al. Toward adaptive deep brain
Funct. Neurosurg. 95, 86–92 (2017). the ventralis intermedius nucleus of the thalamus as a stimulation for dystonia. Neurosurg. Focus 45, E3
41. De Ridder, D., Vanneste, S., Plazier, M., van der Loo, E. treatment of movement disorders. J. Neurosurg. 84, (2018).
& Menovsky, T. Burst spinal cord stimulation: toward 203–214 (1996). 91. Sinclair, N. C. et al. Subthalamic nucleus deep brain
paresthesia-​free pain suppression. Neurosurgery 66, 65. Kirsch, A. D., Hassin-​Baer, S., Matthies, C., stimulation evokes resonant neural activity. Ann.
986–990 (2010). Volkmann, J. & Steigerwald, F. Anodic versus Neurol. 83, 1027–1031 (2018).
42. Kapural, L. et al. Novel 10-kHz high-​frequency therapy cathodic neurostimulation of the subthalamic 92. Rosa, M. et al. Adaptive deep brain stimulation
(HF10 therapy) is superior to traditional low-​frequency nucleus: A randomized-​controlled study of acute controls levodopa-​induced side effects in Parkinsonian
spinal cord stimulation for the treatment of chronic clinical effects. Parkinsonism Relat. Disord. 55, patients. Mov. Disord. 32, 628–629 (2017).
back and leg pain: the SENZA-​RCT randomized 61–67 (2018). 93. Deffains, M., Iskhakova, L., Katabi, S., Israel, Z. &
controlled trial. Anesthesiology 123, 851–860 66. Grill, W. M. Temporal pattern of electrical stimulation Bergman, H. Longer β oscillatory episodes reliably
(2015). is a new dimension of therapeutic innovation. identify pathological subthalamic activity in
43. Schultz, D. M. et al. Sensor-​driven position-​adaptive Curr. Opin. Biomed. Eng. 8, 1–6 (2018). Parkinsonism. Mov. Disord. 33, 1609–1618 (2018).
spinal cord stimulation for chronic pain. Pain. Physician 67. Brocker, D. T. et al. Optimized temporal pattern 94. Little, S. et al. Bilateral adaptive deep brain stimulation
15, 1–12 (2012). of brain stimulation designed by computational is effective in Parkinson’s disease. J. Neurol. Neurosurg.
44. Hosain, M. K., Kouzani, A. Z., Tye, S. J., Abulseoud, O. A. evolution. Sci. Transl. Med. 9, eaah3532 (2017). Psychiatry 87, 717–721 (2016).
& Berk, M. Design and analysis of an antenna for 68. Birdno, M. J. et al. Stimulus features underlying 95. Little, S. et al. Adaptive deep brain stimulation for
wireless energy harvesting in a head-​mountable DBS reduced tremor suppression with temporally Parkinson’s disease demonstrates reduced speech
device. Annu. Int. Conf. IEEE Eng. Med. Biol. Soc. 2013, patterned deep brain stimulation. J. Neurophysiol. side effects compared to conventional stimulation in
3078–3081 (2013). 107, 364–383 (2012). the acute setting. J. Neurol. Neurosurg. Psychiatry
45. Hong, B. et al. Detection of bacterial DNA on 69. Brocker, D. T. et al. Improved efficacy of temporally 87, 1388–1389 (2016).
neurostimulation systems in patients without overt non-​regular deep brain stimulation in Parkinson’s 96. Shah, S. A., Tinkhauser, G., Chen, C. C., Little, S.
infection. Clin. Neurol. Neurosurg. 184, 105399 disease. Exp. Neurol. 239, 60–67 (2013). & Brown, P. Parkinsonian tremor detection from
(2019). 70. Krauss, J. K., Yianni, J., Loher, T. J. & Aziz, T. Z. Deep subthalamic nucleus local field potentials for
46. Jitkritsadakul, O. et al. Systematic review of hardware-​ brain stimulation for dystonia. J. Clin. Neurophysiol. closed-loop deep brain stimulation. Annu. Int.
related complications of deep brain stimulation: do 21, 18–30 (2004). Conf. IEEE Eng. Med. Biol. Soc. 2018, 2320–2324
new indications pose an increased risk? Brain Stimul. 71. Cassar, I. R., Titus, N. D. & Grill, W. M. An improved (2018).
10, 967–976 (2017). genetic algorithm for designing optimal temporal 97. Cagnan, H. et al. Stimulating at the right time: phase-​
47. Piacentino, M., Pilleri, M. & Bartolomei, L. Hardware-​ patterns of neural stimulation. J. Neural Eng. 14, specific deep brain stimulation. Brain 140, 132–145
related infections after deep brain stimulation surgery: 066013 (2017). (2017).
review of incidence, severity and management in 72. Lee, S., Asaad, W. F. & Jones, S. R. Computational 98. Basu, I. et al. Pathological tremor prediction using
212 single-​center procedures in the first year after modeling to improve treatments for essential tremor. surface electromyogram and acceleration: potential
implantation. Acta Neurochir. 153, 2337–2341 (2011). Drug Discov. Today Dis. Model. 19, 19–25 (2016). use in ‘ON-​OFF’ demand driven deep brain stimulator
48. Tarakji, K. G. et al. Antibacterial envelope to prevent 73. Tass, P. A. Phase Resetting in Medicine and Biology: design. J. Neural Eng. 10, 036019 (2013).
cardiac implantable device infection. N. Engl. J. Med. Stochastic Modelling and Data Analysis (Springer, 99. Tan, H. et al. Decoding voluntary movements and
380, 1895–1905 (2019). 1999). postural tremor based on thalamic LFPs as a basis for
49. Sauer, T., Wolf, M. E., Blahak, C., Capelle, H. H. & 74. Tass, P. A. A model of desynchronizing deep brain closed-​loop stimulation for essential tremor. Brain
Krauss, J. K. Neuroleptic-​like malignant syndrome stimulation with a demand-​controlled coordinated Stimul. 12, 858–867 (2019).
after battery depletion in a patient with deep brain reset of neural subpopulations. Biol. Cybern. 89, 100. Velisar, A. et al. Dual threshold neural closed loop
stimulation for secondary parkinsonism. Mov. Disord. 81–88 (2003). deep brain stimulation in Parkinson disease patients.
Clin. Pract. 4, 629–631 (2017). 75. Popovych, O. V. & Tass, P. A. Control of abnormal Brain Stimul. 12, 868–876 (2019).
50. Hancu, I. et al. On the (Non-)equivalency of monopolar synchronization in neurological disorders. Front. 101. Morrell, M. J. & RNS System in Epilepsy Study Group.
and bipolar settings for deep brain stimulation fMRI Neurol. 5, 268 (2014). Responsive cortical stimulation for the treatment of
studies of Parkinson’s disease patients. J. Magn. 76. Markram, H., Lubke, J., Frotscher, M. & Sakmann, B. medically intractable partial epilepsy. Neurology 77,
Reson. Imaging 49, 1736–1749 (2019). Regulation of synaptic efficacy by coincidence of 1295–1304 (2011).
51. Bronstein, J. M. et al. The rationale driving the postsynaptic APs and EPSPs. Science 275, 213–215 102. Elder, C., Friedman, D., Devinsky, O., Doyle, W.
evolution of deep brain stimulation to constant-​current (1997). & Dugan, P. Responsive neurostimulation targeting
devices. Neuromodulation 18, 85–88 (2015). 77. Tass, P. A. & Majtanik, M. Long-​term anti-​kindling the anterior nucleus of the thalamus in 3 patients with
52. Lettieri, C. et al. Clinical outcome of deep brain effects of desynchronizing brain stimulation: a treatment-​resistant multifocal epilepsy. Epilepsia
stimulation for dystonia: constant-​current or constant-​ theoretical study. Biol. Cybern. 94, 58–66 (2006). Open. 4, 187–192 (2019).
voltage stimulation? A non-​randomized study. Eur. J. 78. Hauptmann, C. & Tass, P. A. Cumulative and 103. Voges, B. R. et al. Deep brain stimulation of anterior
Neurol. 22, 919–926 (2015). after-effects of short and weak coordinated reset nucleus thalami disrupts sleep in epilepsy patients.
53. Preda, F. et al. Switching from constant voltage to stimulation: a modeling study. J. Neural Eng. 6, Epilepsia 56, e99–e103 (2015).
constant current in deep brain stimulation: a 016004 (2009). 104. Boon, P. et al. A prospective, multicenter study of
multicenter experience of mixed implants for movement 79. Tass, P. A. et al. Coordinated reset has sustained cardiac-​based seizure detection to activate vagus
disorders. Eur. J. Neurol. 23, 190–195 (2016). aftereffects in Parkinsonian monkeys. Ann. Neurol. 72, nerve stimulation. Seizure 32, 52–61 (2015).
54. Lempka, S. F., Johnson, M. D., Miocinovic, S., 816–820 (2012). 105. Fisher, R. S. et al. Automatic vagus nerve stimulation
Vitek, J. L. & McIntyre, C. C. Current-​controlled deep 80. Adamchic, I. et al. Coordinated reset neuromodulation triggered by ictal tachycardia: clinical outcomes and
brain stimulation reduces in vivo voltage fluctuations for Parkinson’s disease: proof-​of-concept study. Mov. device performance — the U.S. E-37 Trial.
observed during voltage-​controlled stimulation. Disord. 29, 1679–1684 (2014). Neuromodulation 19, 188–195 (2016).
Clin. Neurophysiol. 121, 2128–2133 (2010). 81. Wang, J. et al. Coordinated reset deep brain 106. Wolf, M. E., Blahak, C., Saryyeva, A., Schrader, C.
55. Cheung, T. et al. Longitudinal impedance variability stimulation of subthalamic nucleus produces long-​ & Krauss, J. K. Deep brain stimulation for dystonia-​
in patients with chronically implanted DBS devices. lasting, dose-​dependent motor improvements in the choreoathetosis in cerebral palsy: pallidal versus
Brain Stimul. 6, 746–751 (2013). 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine non-​ thalamic stimulation. Parkinsonism Relat. Disord. 63,
56. Grill, W. M. Model-​based analysis and design human primate model of parkinsonism. Brain Stimul. 209–212 (2019).
of waveforms for efficient neural stimulation. 9, 609–617 (2016). 107. Sani, O. G. et al. Mood variations decoded from multi-​
Prog. Brain Res. 222, 147–162 (2015). 82. Bouthour, W. et al. Biomarkers for closed-​loop deep site intracranial human brain activity. Nat. Biotechnol.
57. Akbar, U. et al. Randomized, blinded pilot testing of brain stimulation in Parkinson disease and beyond. 36, 954–961 (2018).
nonconventional stimulation patterns and shapes in Nat. Rev. Neurol. 15, 343–352 (2019). 108. Kremen, V. et al. Integrating brain implants with local
Parkinson’s disease and essential tremor: evidence for 83. Hoang, K. B. & Turner, D. A. The emerging role of and distributed computing devices: a next generation
further evaluating narrow and biphasic pulses. biomarkers in adaptive modulation of clinical brain epilepsy management system. IEEE J. Transl. Eng.
Neuromodulation 19, 343–356 (2016). stimulation. Neurosurgery 85, E430–E439 (2019). Health Med. 6, 2500112 (2018).
58. De Jesus, S. et al. Square biphasic pulse deep brain 84. Shute, J. B. et al. Thalamocortical network activity 109. Khanna, P. et al. Enabling closed-loop
stimulation for essential tremor: the BiP tremor study. enables chronic tic detection in humans with Tourette neurostimulation research with downloadable
Parkinsonism Relat. Disord. 46, 41–46 (2018). syndrome. Neuroimage Clin. 12, 165–172 (2016). firmware upgrades. IEEE Biomed. Circuits Syst. Conf.
59. McIntyre, C. C. & Grill, W. M. Selective microstimulation 85. Swann, N. C. et al. Adaptive deep brain stimulation https://doi.org/10.1109/BioCAS.2015.7348348
of central nervous system neurons. Ann. Biomed. Eng. for Parkinson’s disease using motor cortex sensing. (2015).
28, 219–233 (2000). J. Neural Eng. 15, 046006 (2018). 110. Liu, T. et al. Improved subthalamic nucleus depiction
60. Hofmann, L., Ebert, M., Tass, P. A. & Hauptmann, C. 86. Herron, J. A. et al. Chronic electrocorticography with quantitative susceptibility mapping. Radiology
Modified pulse shapes for effective neural stimulation. for sensing movement intention and closed-​loop 269, 216–223 (2013).
Front. Neuroeng. 4, 9 (2011). deep brain stimulation with wearable sensors in 111. Wang, Y. & Liu, T. Quantitative susceptibility mapping
61. Popovych, O. V., Lysyansky, B., Rosenblum, M., an essential tremor patient. J. Neurosurg. 127, (QSM): decoding MRI data for a tissue magnetic
Pikovsky, A. & Tass, P. A. Pulsatile desynchronizing 580–587 (2017). biomarker. Magn. Reson. Med. 73, 82–101 (2015).

www.nature.com/nrneurol
Reviews

112. Sudhyadhom, A., Haq, I. U., Foote, K. D., Okun, M. S. 130. Chaturvedi, A., Lujan, J. L. & McIntyre, C. C. Artificial used for deep brain stimulation? Neurosurgery 57,
& Bova, F. J. A high resolution and high contrast MRI neural network based characterization of the volume 1056–1062 (2005).
for differentiation of subcortical structures for DBS of tissue activated during deep brain stimulation. 151. Boutet, A. et al. 3-Tesla MRI of deep brain stimulation
targeting: the Fast Gray Matter Acquisition T1 J. Neural Eng. 10, 056023 (2013). patients: safety assessment of coils and pulse
Inversion Recovery (FGATIR). Neuroimage 47 131. Schmidt, C., Grant, P., Lowery, M. & van Rienen, U. sequences. J. Neurosurg. 132, 586–594 (2019).
(Suppl. 2), T44–T52 (2009). Influence of uncertainties in the material properties 152. Boutet, A. et al. Functional MRI safety and artifacts
113. Horn, A. et al. Lead-​DBS v2: towards a comprehensive of brain tissue on the probabilistic volume of tissue during deep brain stimulation: experience in 102
pipeline for deep brain stimulation imaging. activated. IEEE Trans. Biomed. Eng. 60, 1378–1387 patients. Radiology 293, 174–183 (2019).
Neuroimage 184, 293–316 (2019). (2013). 153. Denning, T., Matsuoka, Y. & Kohno, T. Neurosecurity:
114. Coenen, V. A., Madler, B., Schiffbauer, H., Urbach, H. & 132. Butson, C. R., Cooper, S. E., Henderson, J. M. & security and privacy for neural devices. Neurosurg.
Allert, N. Individual fiber anatomy of the subthalamic McIntyre, C. C. Patient-​specific analysis of the volume Focus. 27, E7 (2009).
region revealed with diffusion tensor imaging: of tissue activated during deep brain stimulation. 154. Zizek, S. Like a Thief in Broad Daylight — Power in the
a concept to identify the deep brain stimulation Neuroimage 34, 661–670 (2007). Era of Post-​human Capitalism (Seven Stories Press,
target for tremor suppression. Neurosurgery 68, 133. Horn, A. et al. Deep brain stimulation induced 2018).
1069–1075 (2011). normalization of the human functional connectome in 155. Hittinger, E. & Jaramillo, P. Internet of Things:
115. Tourdias, T., Saranathan, M., Levesque, I. R., Su, J. Parkinson’s disease. Brain 142, 3129–3143 (2019). energy boon or bane? Science 364, 326–328
& Rutt, B. K. Visualization of intra-​thalamic nuclei 134. Akram, H. et al. Subthalamic deep brain stimulation (2019).
with optimized white-​matter-nulled MPRAGE at 7T. sweet spots and hyperdirect cortical connectivity in 156. [No authors listed] A connected world will be a
Neuroimage 84, 534–545 (2014). Parkinson’s disease. Neuroimage 158, 332–345 playground for hackers. The Economist https://
116. Kanowski, M. et al. Direct visualization of anatomic (2017). www.economist.com/technology-​quarterly/2019/09/
subfields within the superior aspect of the human lateral 135. Bot, M. et al. Deep brain stimulation for Parkinson’s 12/a-​connected-world-​will-be-​a-playground-​for-hackers
thalamus by MRI at 7T. AJNR Am. J. Neuroradiol. 35, disease: defining the optimal location within the (2019).
1721–1727 (2014). subthalamic nucleus. J. Neurol. Neurosurg. Psychiatry 157. Pugh, J., Pycroft, L., Sandberg, A., Aziz, T. &
117. Duchin, Y. et al. Patient-​specific anatomical model for 89, 493–498 (2018). Savulescu, J. Brainjacking in deep brain stimulation
deep brain stimulation based on 7 Tesla MRI. PLoS 136. Dembek, T. A. et al. Probabilistic sweet spots and autonomy. Ethics Inf. Technol. 20, 219–232
One 13, e0201469 (2018). predict motor outcome for deep brain stimulation in (2018).
118. Plantinga, B. R. et al. Individualized parcellation of the Parkinson disease. Ann. Neurol. 86, 527–538 (2019). 158. Spiegel, E. A., Wycis, H. T., Marks, M. & Lee, A. J.
subthalamic nucleus in patients with Parkinson’s 137. Horn, A. et al. Connectivity predicts deep brain Stereotaxic apparatus for operations on the human
disease with 7T MRI. Neuroimage 168, 403–411 stimulation outcome in Parkinson disease. Ann. brain. Science 106, 349–350 (1947).
(2018). Neurol. 82, 67–78 (2017). 159. Delgado, J. M. R. Physical Control of the Mind: Toward
119. Dembek, T. A. et al. Directional DBS leads show large 138. Neumann, W. J. et al. A localized pallidal physiomarker a Psychocivilized Society (Harper and Row, 1969).
deviations from their intended implantation orientation. in cervical dystonia. Ann. Neurol. 82, 912–924 (2017).
Parkinsonism Relat. Disord. 67, 117–121 (2019). 139. Reich, M. M. et al. Probabilistic mapping of the Acknowledgements
120. Bonmassar, G., Angelone, L. M. & Makris, N. A virtual antidystonic effect of pallidal neurostimulation: a The work on this manuscript was supported by an uncondi-
patient simulator based on human connectome and multicentre imaging study. Brain 142, 1386–1398 tional grant of the World Society for Stereotactic and
7 T MRI for deep brain stimulation. Int. J. Adv. Life Sci. (2019). Functional Neurosurgery (WSSFN). The working process was
6, 364–372 (2014). 140. Schonecker, T. et al. Postoperative MRI localisation of coordinated with the Research and Education committees of
121. Husch, A., Petersen, M. V., Gemmar, P., Goncalves, J. electrodes and clinical efficacy of pallidal deep brain the WSSFN.
& Hertel, F. PaCER — a fully automated method for stimulation in cervical dystonia. J. Neurol. Neurosurg.
electrode trajectory and contact reconstruction in Psychiatry 86, 833–839 (2015). Author contributions
deep brain stimulation. Neuroimage Clin. 17, 80–89 141. Al-​Fatly, B. et al. Connectivity profile of thalamic deep All authors contributed to all aspects of manuscript
(2017). brain stimulation to effectively treat essential tremor. preparation.
122. Lauro, P. M. et al. DBSproc: an open source process Brain 142, 3086–3098 (2019).
for DBS electrode localization and tractographic 142. Dembek, T. A. et al. Probabilistic mapping of deep Competing interests
analysis. Hum. Brain Mapp. 37, 422–433 (2016). brain stimulation effects in essential tremor. J. K. K. is a consultant for Medtronic and Boston Scientific. P. B.
123. Miocinovic, S., Noecker, A. M., Maks, C. B., Neuroimage Clin. 13, 164–173 (2017). is a consultant for Medtronic. W. M. G. is the Director, Chief
Butson, C. R. & McIntyre, C. C. Cicerone: stereotactic 143. Baldermann, J. C. et al. Connectivity profile predictive Scientific Officer and share owner of Deep Brain Innovations,
neurophysiological recording and deep brain stimulation of effective deep brain stimulation in obsessive– LLC. He also receives royalty payments for licensed patents on
electrode placement software system. Acta Neurochir. compulsive disorder. Biol. Psychiatry 85, 735–743 temporal patterns of deep brain stimulation. M. I. H. has
Suppl. 97, 561–567 (2007). (2019). received travel expenses and honoraria from Boston Scientific
124. Horn, A. & Kuhn, A. A. Lead-​DBS: a toolbox for deep 144. Horn, A. The impact of modern-​day neuroimaging on for speaking at meetings. A. H. was supported by the German
brain stimulation electrode localizations and the field of deep brain stimulation. Curr. Opin. Neurol. Research Council (DFG grant 410169619) and reports lecture
visualizations. Neuroimage 107, 127–135 (2015). 32, 511–520 (2019). fees from Medtronic and Boston Scientific unrelated to the
125. Milchenko, M. et al. ESM-​CT: a precise method for 145. Horn, A. et al. Probabilistic conversion of neurosurgical present work. P. A. T. works as a consultant for Boston Scientific
localization of DBS electrodes in CT images. DBS electrode coordinates into MNI space. Neuroimage Neuromodulation. J. V. works as a consultant to Boston
J. Neurosci. Methods 308, 366–376 (2018). 150, 395–404 (2017). Scientific, Medtronic, and Newronika and has received hono-
126. Chakravorti, S. et al. Validation of an automatic 146. Lozano, A. M. & Lipsman, N. Probing and regulating raria for lectures from Boston Scientific and Medtronic as well
algorithm to identify NeuroPace depth leads in CT dysfunctional circuits using deep brain stimulation. as research grants from Boston Scientific and Medtronic. A. M.
images. Proc. SPIE https://doi.org/10.1117/ Neuron 77, 406–424 (2013). L. has served as a consultant for Boston Scientific, Medtronic,
12.2512580 (2019). 147. Choi, K. S., Riva-​Posse, P., Gross, R. E. & Mayberg, H. S. Aleva, and Abbott and is a co-​f ounder of Functional
127. Sitz, A. et al. Determining the orientation angle of Mapping the “depression switch” during intraoperative Neuromodulation. All other authors declare no competing
directional leads for deep brain stimulation using testing of subcallosal cingulate deep brain stimulation. interests.
computed tomography and digital x-​ray imaging: JAMA Neurol. 72, 1252–1260 (2015).
a phantom study. Med. Phys. 44, 4463–4473 148. Riva-​Posse, P. et al. A connectomic approach for Peer review information
(2017). subcallosal cingulate deep brain stimulation surgery: Nature Reviews Neurology thanks V. Visser-​Vandewalle and
128. Boutet, A. et al. Neuroimaging technological prospective targeting in treatment-​resistant Y. Temel for their contribution to the peer review of this work.
advancements for targeting in functional neurosurgery. depression. Mol. Psychiatry 23, 843–849 (2018).
Curr. Neurol. Neurosci. Rep. 19, 42 (2019). 149. Li, N. et al. A unified connectomic target for deep Publisher’s note
129. Ewert, S. et al. Optimization and comparative brain stimulation in obsessive-​compulsive disorder. Springer Nature remains neutral with regard to jurisdictional
evaluation of nonlinear deformation algorithms for Nat. Commun. 11, 3364 (2020). claims in published maps and institutional affiliations.
atlas-​based segmentation of DBS target nuclei. 150. Rezai, A. R. et al. Is magnetic resonance imaging
Neuroimage 184, 586–598 (2019). safe for patients with neurostimulation systems © Springer Nature Limited 2020

Nature Reviews | Neurology

You might also like