You are on page 1of 15

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/332783714

The use of transcranial magnetic stimulation as a treatment for movement


disorders: A critical review

Article  in  Movement Disorders · April 2019


DOI: 10.1002/mds.27705

CITATIONS READS

0 421

5 authors, including:

Anna Latorre Lorenzo Rocchi


University College London University College London
42 PUBLICATIONS   217 CITATIONS    58 PUBLICATIONS   412 CITATIONS   

SEE PROFILE SEE PROFILE

Alfredo Berardelli Kailash Bhatia


Sapienza University of Rome University College London
625 PUBLICATIONS   24,821 CITATIONS    873 PUBLICATIONS   30,294 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

muscle fatigue and SICI View project

role of altered temporal discrimination of somatosensory input in the pathophysiology in movement disorders View project

All content following this page was uploaded by Anna Latorre on 03 May 2019.

The user has requested enhancement of the downloaded file.


REVIEW

The Use of Transcranial Magnetic Stimulation as a Treatment


for Movement Disorders: A Critical Review
Anna Latorre, MD,1,2 Lorenzo Rocchi, MD,1 Alfredo Berardelli, MD,2,3
Kailash P. Bhatia, FRCP, MD,1 and John C. Rothwell, PhD1*

1
Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology University College London, London, UK
2
Department of Human Neurosciences, Sapienza University of Rome, Rome, Italy
3
IRCCS Neuromed Institute, Pozzilli, Isernia, Italy

A B S T R A C T : Background: Transcranial magnetic stim- and possible mechanistic explanations for the relatively
ulation is a safe and painless non-invasive brain stimula- minor effects of transcranial magnetic stimulation are dis-
tion technique that has been largely used in the past cussed. Possible ways to improve the methodology and
30 years to explore cortical function in healthy participants achieve greater therapeutic efficacy are discussed.
and, inter alia, the pathophysiology of movement disor- Conclusion: Despite the promising and robust rationales
ders. During the years, its use has evolved from primarily for the use of transcranial magnetic stimulations as a treat-
research purposes to treatment of a large variety of ment tool in movement disorders, the results taken as a
neurological and psychiatric diseases. In this article, we whole are not as successful as were initially expected.
illustrate the basic principles on which the therapeutic There is encouraging evidence that transcranial magnetic
use of transcranial magnetic stimulation is based and stimulation may improve motor symptoms and depression
review the clinical trials that have been performed in in Parkinson’s disease, but the efficacy in other movement
patients with movement disorders. disorders is unclear. Possible improvements in methodol-
Methods: A search of the PubMed database for research ogy are on the horizon but have yet to be implemented in
and review articles was performed on therapeutic appli- large clinical studies. © 2019 International Parkinson and
cations of transcranial magnetic stimulation in movement Movement Disorder Society
disorders. The search included the following conditions:
Parkinson’s disease, dystonia, Tourette syndrome and Key Words: movement disorders; neurophysiology; plas-
other chronic tic disorders, Huntington’s disease and ticity; repetitive transcranial magnetic stimulation; trans-
choreas, and essential tremor. The results of the studies cranial magnetic stimulation

What Is Transcranial Magnetic been largely used to explore cortical function in healthy
subjects and in patients with neurological and psychiat-
Stimulation? ric disorders, both for experimental and clinical purposes.
Transcranial magnetic stimulation (TMS) is a nonin- Most of this work has been performed on the human
primary motor cortex (M1) and has given important
vasive neurophysiological technique that can stimulate
pathophysiological and clinical insights in the field of
the human brain through the intact skull without pro-
movement disorders (MD).2-4
ducing significant discomfort. TMS was developed in
Magnetic stimulators consists of capacitors able to dis-
1985 by Barker and colleagues,1 and since then it has
charge a large current (with a peak of amplitude of up
-*Correspondence
- - - - - - - - - - - - - - -to:- - Dr.
- - - John
- - - - -C.- -Rothwell,
- - - - - - - 33
- - -Queen
- - - - - -Square,
- - - - - - London
------- to 10000 A) that lasts about 1 millisecond and flows
WC1N 3BG, UK; E-mail: j.rothwell@ucl.ac.uk through an induction coil placed on the scalp.5 This cur-
Relevant conflicts of interests/financial disclosures: Nothing to rent produces a strong magnetic field, perpendicular to
report. the coil, which can reach values of up to 3 Tesla; it easily
Received: 16 December 2018; Revised: 4 April 2019; Accepted: passes through the scalp and skull and induces a second-
7 April 2019 ary electric (“eddy”) current in the brain.2 The intensity
Published online 00 Month 2019 in Wiley Online Library
of the magnetic field declines quickly with distance from
(wileyonlinelibrary.com). DOI: 10.1002/mds.27705 the coil so that it is usually assumed that neural stimulation

Movement Disorders, 2019 1


L A T O R R E E T A L

is limited to the cortex and superficial subcortical white In all of these cases, it is assumed that therapeutic
matter.6 Stimulation over M1 can produce descending effects depend on changes in synaptic plasticity pro-
activity in the corticospinal pathway that activates muscles duced by periods of rTMS.
on the opposite side of the body and produces a visible
twitch that is easily measured using surface electromyogra-
phy.7 This is known as a motor evoked potential (MEP).
TMS and Synaptic Plasticity
In addition to a single pulse, TMS pairs of pulses can be The data supporting rTMS effects on synaptic plastic-
delivered through the same coil to study intracortical ity in the brain mostly come from TMS studies of M1.
inhibitory and excitatory circuits.8-12 TMS can also assess Depending on the intensity and direction of the current,
inputs to M1 from other areas by observing how the TMS can activate the large corticospinal neurones that
constitute the corticospinal (or pyramidal) tract either
MEP is modulated by a preceding stimulus, the latter
directly or trans-synaptically. In the second case, the
being either a magnetic pulse applied over a different
TMS pulse first discharges an unknown set of neurons
cortical area13 or a stimulus activating afferents from var-
that secondarily activate the corticospinal neurones via
ious sensory modalities.14-16 It is also possible to apply
excitatory synapses.2,29 Therefore, when a TMS pulse
repetitive TMS (rTMS) in the form of short17,18 or long is given, activity at these synapses causes discharge of
trains, either regular or patterned,19-24 or associated with the corticospinal neurones and consequent conduction
diverse cortical or sensory stimulation.14,18,25 These pro- of the impulses to the spinal cord and muscle. It is well
tocols can induce long-term changes in cortical excitabil- known from animal experiments that repetitive stimula-
ity that are the result of changes in synaptic plasticity. tion at the synaptic level can enhance or reduce synap-
The most common patterns of stimulation of rTMS are tic transmission, a phenomenon known as long-term
described in Table 1 and Figure 1.26,27 potentiation (LTP) or long-term depression (LTD). LTP
is often induced by a short period of high-frequency
repetitive stimulations, whereas LTD usually occurs after
low-frequency stimulation.30 Because TMS indirectly acti-
Why rTMS Might Be Used vates synapses, repeated pulses of TMS (ie, rTMS) can in
as a Therapeutic Tool theory activate the same set of synaptic connections multi-
ple times and therefore replicate the situation seen in ani-
The rational use of non-invasive brain stimulation as mal experiments. Thus, rTMS over M1 can lead to
a therapeutic tool is to provide additional benefit to medium-term changes in the amplitude of MEPs that per-
conventional treatment, especially for refractory symp- sists after the end of stimulation. By analogy with the ani-
toms or in patients in whom surgical approaches are mal experiments, high-frequency rTMS increases MEPs,
contraindicated. With this purpose and in view of its whereas low frequency reduces MEPs, reflecting an
safety and lower side effects when compared with elec- increase or decrease of cortical excitability. It is thought
troconvulsive therapy, rTMS was used for the first time that these changes are caused respectively by synaptic
in 1993 to treat drug-resistant major depression disor- LTP/LTD because they are no longer present in individ-
der.28 In light of the successful results, rTMS was then uals pretreated with drugs that interfere with N-methyl-
applied in other psychiatric and neurological condi- D-aspartate receptor (NMDA), receptors (known to be
tions, including MD, as a possible therapy. involved in LTP/LTD processes).27,31
Because the after-effects persist only 30 minutes after
TABLE 1. Commonly used rTMS techniques
repetitive stimulation, they probably represent the early
phase of plasticity (early LTP/LTD) in animal experi-
rTMS: consecutive TMS stimuli at a specific frequency, with variable ments. This is a stage where the change in efficiency is
interstimulus intervals. probably the result of a combination of increased trans-
• Short train of regular 5 Hz rTMS: MEP facilitation mitter release from the presynaptic neurone and an
• Long train of regular ≤1 Hz rTMS: MEP inhibition increase in response to transmitter by the postsynaptic
• Long train of regular ≥5 Hz rTMS: MEP facilitation neurone (eg, by inserting more receptors in the mem-
• Short high-frequency trains (bursts) at a predefined repetition rate- brane). Protein synthesis is required to produce more
TBS: intermittent TBS induces MEP facilitation (LTP-like plasticity),
permanent changes, such as increases in synaptic effi-
continuous TBS-induced MEP inhibition (LTD-like plasticity)
• Paired associative stimulation (PAS): 100-200 pairs of electrical
cacy or the formation of new synapses.30 These conclu-
stimuli at the median nerve at the wrist and TMS stimuli to the sions have been reinforced by experiments in which
contralateral M1 (interval of about 25 milliseconds) are given every rTMS has been applied to the brains of rodents. These
4-10 seconds (LTP-like plasticity) have revealed changes at a molecular level that would
be consistent with the idea that rTMS in humans may
rTMS, repetitive transcranial magnetic stimulation; LTD, long-term depres-
sion; LTP, long-term potentiation; MEP, motor evoked potential; TBS, theta
be able to interact with synaptic plasticity. The data
burst stimulation. reveal structural effects on synaptic morphology32 as

2 Movement Disorders, 2019


R T M S F O R T H E T R E A T M E N T O F M O V E M E N T D I S O R D E R S

FIG. 1. The most common patterns of stimulation of rTMS. cTBS, continuous theta burst stimulation; ES, electrical stimulation; HF, high frequency;
iTBS, intermittent theta burst stimulation; LF, low frequency; MS, magnetic stimulation; PAS, paired associative stimulation; rTMS, repetitive transcranial
magnetic stimulation.

well as changes in Calcium ion (Ca2+) dynamics, which Approaches to Using rTMS as a Therapy
then modulate the expression of glutamate receptors33 The effects of rTMS in humans are transitory and last
and immediate early genes that regulate the expression of about 30 to 60 minutes, depending on parameters such
specific genes.34 as the number of pulses applied, the rate of application,
In addition to rTMS, the effects on synaptic plasticity and the intensity of each stimulus.31 Practically this
can be produced by a second method known as spike- would seem to have a limited therapeutic effect because
timing dependent plasticity. This involves repeated the intent is to induce lasting changes in patients’ symp-
pairing of synaptic inputs to a neurone with a subse- toms. Nevertheless, in animal experiments, it is possible
quent discharge (spike) of the neurone itself. This can to produce more permanent changes in synapses if stim-
be replicated in humans by pairing an input to cortex ulation is repeated after a pause of a few minutes or
produced by electrically stimulating a peripheral nerve hours.40 Repeated induction of plasticity is thought to
(eg, median nerve) with a TMS pulse to the cortex. In reinforce the change from transitory early to more per-
humans this is called paired associative stimulation manent late LTP/LTD. The approach in humans is simi-
(PAS).35 PAS can increase or decrease MEPs for 30 lar, and there is evidence that repeated rTMS
minutes similarly to rTMS. Novel PAS protocols, using administration might produce cumulative effects, which
a cortical–cortical approach or different sensory stimuli, depend on the number of sessions.41-44 In most cases,
have also been applied in humans.14,18,25 rTMS is given daily for 5, 10, or more days.45 For
There are other mechanisms that might mediate the instance, in depression the remission rates are higher
effect of rTMS and are probably independent of synap- with longer rTMS treatment (up to 6 weeks),46,47 and
tic plasticity. For instance, rTMS has been shown to act more recent studies indicate that at least 20 to 30 ses-
on the neuroendocrine system by decreasing corticoste- sions are required to have optimal effects48; in fact,
roid levels in rats36 and humans.37 Other effects include even longer courses might be needed in some cases.49 It
the prevention of neuronal death, as demonstrated by is thought that rTMS can gradually induce changes in
applying low-intensity magnetic stimulation in cultures functional and structural connectivity that normalize
of cortical neurons,38 and increased expression of the the depression-associated network dysfunctions.50
glial fibrillary acidic protein in astrocytes.39 Overall, Because of the bulk of the stimulating coil and its
data on the effects of rTMS beyond synaptic plasticity electrical supply, TMS is usually given when patients
are still scarce, but increasing information in this regard are seated and relaxed. For example, in the treatment
might help to improve the therapeutic role of rTMS. of depression, patients are given rTMS daily when seated

Movement Disorders, 2019 3


L A T O R R E E T A L

in a comfortable chair. A different approach is to give frequency prefrontal rTMS (for a total of 782 subjects
rTMS prior to a training intervention (eg, prior to stan- studied).47,71-73 Of these (291 participants), two revealed
dard physiotherapy if treating stroke patients51-53). The negative results with no difference in the responder rates
rationale is that therapy involves learning, which itself of the real and the sham treatment groups71,73 (differ-
changes the effectiveness of synaptic connections that are ently from the others, one of these studies involved
required to improve performance in the task. Giving adjunctive TMS and medications starting simultaneously
rTMS prior to this training might increase the effective- rather than TMS as the primary treatment or mon-
ness of this process and result in a more highly targeted otherapy71). The other two trials showed positive results,
set of synaptic changes. that is, 3 to 6 weeks of daily (weekdays only) high-
frequency rTMS over the left DLPFC improved clinical
outcomes significantly more than placebo.47,72 In 2008,
Evidence That rTMS Might Be a Useful Therapy the Food and Drug Administration approved the first
The best evidence that rTMS might be practically use- TMS device for therapeutic clinical use in drug-resistant
ful as a therapy comes from its use in patients with depression, and since then several other studies further
drug-resistant major depression disorder. The rationale confirmed its effectiveness in clinical practice.74-77 More-
for using rTMS originates from imaging studies where over, several meta-analyses have reported that the sham-
patients with depression showed reduced activity (eg, controlled evidence base for the use of TMS in depres-
Blood oxygenation level dependent (BOLD) or glucose sion is clinically and statistically significant.78-82 These
metabolism) of the left dorsolateral prefrontal cortex trials led to a positive endorsement by specialty societies
(DLPFC).54 High-frequency rTMS was therefore sup- and technology assessment entities48,83-85 and a wide
posed to increase excitability by strengthening synaptic acceptance of TMS as a treatment in routine clinical
connections, as suggested by the appropriate changes in practice for patients who have not benefited from treat-
activation seen after a course of rTMS in imaging stud- ment with antidepressant medications.
ies.55,56 However, more recently this hypothesis has
been reviewed. Major depression disorder is now con-
sidered a disorder of dysregulated neural networks.57 Rationale for rTMS in Basal Ganglia Disease
Several functional networks have been studied in major The core anatomical and functional impairment in
depression disorder, but two are of particular interest: MD resides in the basal ganglia, which lie too deep to
the anterior cinguloinsular network or salience network be readily stimulated with conventional TMS coils.
and the ventromedial network or reward network.58 However, it is well known that the basal ganglia are
Although the salience network is crucial for cognitive part of a group of parallel closed circuits (the basal
control and response inhibition and involves the ganglia-thalamo-cortico-basal ganglia loop) that origi-
DLFPC, the reward network corresponds to the classic nate in the cerebral cortex, traverse the thalamus, and
reward circuit and involves the ventromedial prefrontal return to their individual sites of origin in the frontal
cortex. These two networks play opposite roles in lobe.86 Thus by analogy to depression, it could be that
behavioral regulation, and it is conceivable that one is even if direct effects of TMS are limited to the cortex,
active when the other is not and vice versa. Major stimulation over an appropriate cortical region that is
depression disorder has been associated to a pattern of part of the basal ganglia circuitry can influence activity
salience network hypoactivity and reward network within these loops and potentially produce clinical ben-
hyperactivity,59-61 and it has been supposed that excit- efit. In confirmation of this idea, functional MRI
atory rTMS intervention targeting the salience network (fMRI) studies in humans show that TMS of the left
by stimulating DLPFC should exert a therapeutic effect dorsal premotor cortex (PMC) and presupplementary
by enhancing cortico-striatal-thalamic connectivity, motor area (SMA) can increase the BOLD signal in the
whereas an intervention to target the reward network striatum and thalamus,87,88 and PET imaging reveals
through the ventromedial prefrontal cortex should that TMS can produce a transient release of dopamine
exert therapeutic effect by reducing cortico-striatal- in the striatum.89,90 Similarly, cortical TMS can sup-
thalamic connectivity.58 Some imaging studies have press beta activity in the subthalamic nucleus (STN),91
confirmed the hypothesis for the salience network.62 and this might be therapeutically relevant in
rTMS treatment also has other effects and can lead to Parkinson’s Disease (PD) because enhanced beta syn-
significant changes in neural activity in fronto-limbic chronization is related to parkinsonian symptoms.92
brain regions,55,63,64 in neurotransmission (not only Data in animal experiments are consistent with this
close to the stimulation site),65-67 in hypothalamic-pitui- notion that interacting with one node of a complex circuit
tary-adrenal axis function,68 and in concentrations of can change activity at some distance from the node. In
neurotrophic factor.69,70 rodent models of PD, optogenetic studies suggest that
There have been four large, multicenter, randomized stimulation of STN afferent projections from the cortex
controlled trials on the antidepressant effect of high- ameliorates bradykinesia and hypokinesia and causes

4 Movement Disorders, 2019


R T M S F O R T H E T R E A T M E N T O F M O V E M E N T D I S O R D E R S

widespread increased glutamatergic activity in the thalamus TABLE 2. Number of participants in rTMS studies to treat
and other brain regions.93 In primates, implanted electrical motor symptoms in PD
stimulation of motor cortex can alleviate MPTP-induced
Number of patients
symptoms of parkinsonism,94 although attempts to repli- Study included in the study
cate this in humans have not been successful.94,95
In summary, rTMS may affect the strength of synap- 1 Pascual-Leone et al, 1994188 6
tic connections in the human cortex. The evidence for 2 Siebner et al, 1999189 12
3 Siebner et al, 200096 10
this comes mainly from experiments in motor cortex, 4 Shimamoto et al, 200197 9
but it is usually assumed that similar effects will be 5 Boylan et al, 2001115 8
observed in all areas of neocortex. In basal ganglia dis- 6 Khedr et al, 200398 36
ease, it is argued that changing the function of cortical 7 Ikeguchi et al, 200399 12
regions directly with rTMS may have secondary effects 8 Okabe et al, 2003100 85
9 Lefaucheur et al, 2004101 12
on connected structures in the basal ganglia–cortex 10 Bornke et al, 2004102 12
loops. This is the basis for attempting to use it thera- 11 Koch et al, 2005123 8
peutically in basal ganglia disease. Finally, previous evi- 12 Brusa et al, 2006190 10
dence from depression suggests that rTMS can be a 13 Khedr et al, 2006118 55
useful therapeutic option. 14 Lomarev et al, 2006191 18
15 del Olmo et al, 2007192 13
16 Khedr et al, 2007114 20
The Use of rTMS for the 17 Wagle-Shukla et al, 2007121 6
18 Filipovic et al, 2009122 10
Treatment of MD 19 Hamada et al, 2008103 98
20 Koch et al, 2009124 10
Methods: Literature Review 21 Benninger et al, 2009193 10
To select studies for inclusion in this review, the 22 Rothkegel et al, 2009116 22
23 Sedlackova et al, 2009104 10
Medline database (via PubMed, a service of the 24 Arias et al, 2010105 18
National Library of Medicine’s National Center for 25 Filipovic et al, 2010113 10
Biotechnology Information; https://www.ncbi.nlm.nih. 26 Kang et al, 2010194 11
gov) was searched for peer-reviewed papers published 27 Kimura et al, 2011106 12
from 1993 to present day using the following terms: 28 Gonzalez-Garcia et al, 2011195 17
29 Benninger et al, 2011133 26
Parkinson’s disease OR dystonia OR Tourette syn- 30 Benninger et al, 2012107 26
drome OR tics OR Huntington’s disease OR chorea 31 Mak et al, 2013196 22
OR essential tremor AND repetitive transcranial mag- 32 Shirota et al, 2013108 106
netic stimulation OR rTMS OR repetitive TMS OR 33 Maruo et al, 2013109 21
treatment TMS. All types of original articles were 34 Sayin et al, 2014125 17
35 Bologna et al, 2015130 13
included if rTMS protocols were performed for thera- 36 Cerasa et al, 2015126 11
peutic purposes, whereas pure pathophysiological stud- 37 Kim et al, 2015127 17
ies were excluded. Only articles written in English were 38 Tard et al, 2016129 15
included, whereas studies using other forms of non- 39 Brys et al, 2016110 50
invasive brain stimulation techniques were excluded. 40 Mally et al, 2017111 66
41 Kim et al, 2018128 12
Review articles (including meta-analysis) were checked 42 Yokoe et al, 2018112 19
to include relevant articles and information not inde- Total 961
xed in the electronic database.
rTMS, repetitive transcranial magnetic stimulation.

Parkinson’s Disease
Motor Symptoms. Several rTMS studies have been con- postural instability, and gait. Alongside the UPDRS,
ducted with the objective of improving motor symptoms in some of the studies also evaluated motor planning and
PD (Table 2); overall they provided mixed results (for a walking tests.98,99,101,104,105,107,109,111,115-117
review, see ref. 45). These studies differed with regard to The results from three large randomized controlled
rTMS protocols (high/low frequency), cortical areas stimu- trials have shown reasonable evidence that 1 Hz and
lated (M1, prefrontal cortex), sample size, disease duration, 5 Hz rTMS over SMA significantly improves UPDRS
and therapeutic aims (bradykinesia, handwriting, tremor, motor scores (the former stimulation being more effec-
gait, etc.). The degree and type of motor signs that improve tive on posture and gait, whereas the latter had greatest
varies among studies. The main outcome used is the motor effect on bradykinesia) and that rTMS is a promising
part of the Unified Parkinson’s Disease Rating Scale add-on therapy for motor symptoms of PD. In contrast,
(UPDRS),96-114 assessing the overall improvement of the a slower stimulation frequency (0.2 Hz) seems to have
motor symptoms, including bradykinesia, tremor, rigidity, beneficial effects similar to sham stimulation.100,103,108

Movement Disorders, 2019 5


L A T O R R E E T A L

Other cortical targets have also proved successful. One Highlights. According to the current evidence-based
large, double-blind, sham-controlled, randomized, parallel- guidelines on the therapeutic use of rTMS, the publi-
group, fixed-dose study provides class I evidence that in shed data suggest a possible therapeutic potential for
patients with PD on antiparkinsonian treatment, high- motor symptoms in PD; however, evidence is not strong
frequency rTMS (10 Hz) over M1 leads to improvement enough to use rTMS as a routine treatment.45 Indeed,
in overall motor UPDRS and in particular in rigidity the effectiveness of rTMS in PD should be increased by
and bradykinesia, but not in tremor, gait, or axial symp- finding optimal stimulation parameters. In contrast,
toms.110 The effectiveness of high-frequency (5 Hz, high-frequency rTMS of the left DLPFC has a level B
10 Hz and 25 Hz) rTMS over M1 has also been demon- recommendation (“probable efficacy”) in the treatment
strated in other studies on patients not receiving anti- of depression associated with PD.45
parkinsonian medications.98,114,118 Overall, these data
support the efficacy of rTMS in treating motor symptoms Dystonia
in PD, as confirmed by meta-analyses119,120; however, it
The following three principal mechanisms are
remains unclear whether SMA or M1 is the best target,
thought to play a role in the pathophysiology of dysto-
and the results suggest that different rTMS targets might
nia: loss of inhibition at different levels of the central
respond to different frequencies. The remaining studies
nervous system, abnormal sensorimotor integration,
(Table 2) showed variable results, possibly because of
and excessive plasticity.139 Based on this concept, the
the small number of patients included and other con-
rationale for the use of rTMS as a therapeutic tool for
founding factors such as the use of non-realistic sham or
dystonia is to reverse the aforementioned abnormalities,
the different pharmacological state of the patients, which
in particular to act on the inhibitory circuits and possi-
might have affected the UPDRS motor scores.119,120
bly reducing the excessive cortical plasticity.
rTMS has also been evaluated as a possible therapeu-
The following two rTMS protocols have been
tic tool for levodopa-induced dyskinesia, showing some
explored as possible treatments for dystonia with the
positive results when applied on M1 and SMA at low
aim of increasing intracortical inhibition: low-frequency
frequency and on the cerebellum and inferior frontal
rTMS and cTBS. They have been applied on M1 and the
cortex using continuous theta burst stimulation (cTBS)
PMC, SMA, primary somatosensory cortex, and cerebel-
protocol.121-126 Another variable assessed has been
lum with different protocols in patients with writer’s
freezing of gait using high-frequency rTMS over M1
cramp and craniocervical dystonia. Some of the studies
and on SMA and intermittent theta burst stimulation
showed short-lasting objective or subjective improve-
(iTBS) over the premotor cortex,127-129 with contrasting
ment of dystonia, whereas others did not (for a review,
results. In one study, 1 session of cerebellar cTBS did
see ref. 140). On the other hand, iTBS, an excitatory
not improve rest tremor in PD.130
protocol, has been demonstrated in one small study to
Importantly, all of the studies showed no major
be successful in improving cervical dystonic symptoms
adverse effects. In addition, they suggest that different
when applied on the cerebellum.141
protocols have preferential effects on certain symp-
toms.101 Overall, in the majority of the studies, a greater
Highlights. At present, there is insufficient evidence to
rTMS effect was positively associated with a higher
recommend rTMS as a therapeutic tool for dystonia,
number of pulses across sessions and with the total num-
even though some promising results have been publi-
ber of pulses across sessions adjusted by intensity (ie,
shed.140 Physiological data suggested that inhibition of
total number of pulses across sessions times intensity).119
the premotor–motor interactions through a cTBS proto-
col might be the promising strategy for dystonia treat-
Non-motor Symptoms. Among the many non-motor
ment.142 The dorsal PMC seems to be the ideal target
symptoms seen in PD, speech difficulty, cognitive dys-
area for this.45,140
function, and depression are the most frequently stud-
ied in rTMS trials. rTMS is an approved therapy in
several countries for refractory major depression disor- Tourette Syndrome and Other Chronic Tic
der, and high-frequency rTMS of the DLPFC may offer Disorders
a therapeutic option for depression in PD45; in this con- The SMA is involved in the preparation and organiza-
text, rTMS has been shown to have similar efficacy tion of self-initiated movements and has been hypothe-
to antidepressants.131,132 In one study, iTBS applied sized to have an important role in the pathophysiology of
over M1 and DLPFC improved mood, but not motor tics.143,144 Neuroimaging studies have identified the SMA
symptoms in PD.133 It is important to mention that as one of the structures that are hyperactive in Tourette
among the randomized controlled trials that have syndrome,145,146 and this has led to the hypothesis that
assessed the efficacy of rTMS in treating depression in normalizing its excitability might be a potential treatment
PD,134-138 one showed no benefit of left DLPFC rTMS for tics. Low-frequency rTMS of SMA, but not of M1 or
for mood symptoms.110 the PMC, has been shown to reduce tic severity as well as

6 Movement Disorders, 2019


R T M S F O R T H E T R E A T M E N T O F M O V E M E N T D I S O R D E R S

comorbid obsessive-compulsive disorder.147-149 Neverthe- is generated by a dysfunction in the cerebello-thalamo-


less, the results were obtained from open-label studies, cortical network.159
and two more recent double-blind studies did not show a To suppress tremor in ET, rTMS of the cerebellum has
clear significant tic improvement beyond a placebo been studied with variable results. Some improvement in
effect.150,151 In one of these double-blind studies, no dif- tremor rating scales was transiently observed after a sin-
ference was found between real and sham rTMS after gle session of 1 Hz cerebellar rTMS and for 3 weeks
3 weeks of treatment; however, after 3 more weeks of after 5 days of consecutive sessions of 1 Hz rTMS.160,161
open-label active stimulation, participants that were previ- In the first study, the target was 2 centimeters below
ously assigned to the real arm showed a significant reduc- the inion, and in the second it was approximately over
tion in tic severity when compared with baseline.150 the cerebellar lobule VIII. A single session of cerebellar
cTBS did not show modification of tremor frequency,
Highlights. Low-frequency rTMS over SMA has shown smoothness of reaching movements, or clinical rating of
favorable results in reducing tic severity and psychiatric tremor.162 Other targets have been stimulated in further
comorbidity (obsessive-compulsive disorder) in open- studies, such as M1, through a single session of cTBS,163
label studies. However, the lack of significant effects in and pre-SMA, with 15 daily sessions of 1 Hz rTMS.164
double-blind trials questions the possible efficacy of The results on clinical outcomes were null, but cTBS on
the intervention. Larger and appropriately designed M1 could improve tremor measured with an accelerome-
studies to assess rTMS efficacy in treating tic disorders ter.163 Interestingly, recent studies have shown that deliv-
are needed. ering DBS pulses at particular phases of the tremor cycle
causes clinically significant tremor relief,165,166 but this
approach has not been tried yet with noninvasive brain
Huntington’s Disease and Other Choreas stimulation.
To date, only two studies have investigated the effective-
ness of rTMS in Huntington’s disease (HD), giving oppo- Highlights. Although the cerebellum seems a plausible
site results. Brusa and colleagues152 found a significant target in ET, it remains unclear which rTMS protocol
decrease of the involuntary movements after 1 Hz rTMS might be optimal for tremor suppression. The results are
over the SMA, but not after 5 Hz or sham stimulation, in inconclusive so far, and more studies are needed.
4 patients. This result has not been confirmed in 2 other
patients with HD.153 In HD, SMA is thought to play a cen-
tral role in maintaining the executive aspects of motor
Why Didn’t rTMS Induce Clear
control that increases with approaching disease onset.154 Therapeutic Benefits in MD So Far?
The effect of rTMS in HD has also been evaluated on Challenges and Future Directions
non-motor symptoms, such as depression, showing
sustained improvement in mood after 1 Hz rTMS on M1, Despite the promising and robust rationales for the use
but no effect in the 10 Hz or sham rTMS condition.155 of rTMS as a treatment tool in MD, the results are not as
Considering other forms of chorea, cTBS has been tested successful as were expected. However, this does not
for therapeutic purpose in 1 patient with hemichorea extend to all MD and symptoms. The evidence of the
because of hemorrhage of the STN or the substantia nigra efficacy of rTMS in treating motor symptoms as an
or both of these structures. As a result, one session of real add-on therapy to pharmacological treatment in PD is
cTBS produced a dramatic improvement in dyskinesias reasonable,103,108 but despite this it has not had the same
during functional tasks, whereas a sham session did impact as, for example, in major depression disorder.
not.156 There might be several explanations for this. The first is
the lack of a clear rationale to stimulate one cortical area
Highlights. The SMA might be a promising target for compared with another. For instance, SMA and M1
rTMS treatment157; however, the preliminary and con- stimulation have both been reported to be effective,103,108
troversial results need to be confirmed in a large cohort but it remains unclear whether one target is preferable
of patients with HD. rTMS needs to be tested in other than the other or which is the best frequency of rTMS to
forms of chorea to prove its efficacy. use at each site. The matter is different for major depres-
sion disorder because most studies have focused on a sin-
gle recommended site and protocol, producing consistent
Essential Tremor results. Moreover, in contrast to major depression disor-
The pathophysiology of essential tremor (ET) is still der, other more feasible treatment options are available
controversial. The findings are heterogeneous, and they in PD. This might have discouraged the promotion of
possibly reflect the clinical heterogeneity of ET.158 large rTMS trials and the use of rTMS in a hospital set-
However, the evidence of a cerebellar involvement in ting. Therefore, it seems that if a preferred target site
ET is quite robust, and it is very likely that the tremor and protocol can be agreed on, and if there is a clear

Movement Disorders, 2019 7


L A T O R R E E T A L

demonstration that the benefits of rTMS are additional TMS pulses according to different states of cortical excit-
to those of standard therapy in PD, then rTMS might ability. For example, it is known that excitability of M1
become an acceptable treatment. Unfortunately, at the is dependent on the phase, power, and dominant fre-
present time, this is not true for all PD symptoms (ie, quency of ongoing oscillatory activity.177-180 This means
freezing of gait and dyskinesia) and most importantly that if TMS pulses are delivered randomly they are likely
for the other MD. This might be because of the heteroge- to activate populations of neurones in different func-
neity of the protocols employed and of the findings as tional states. Consequently, attempts have been made
well as the lack of any large-scale trials. to tune TMS pulses to different brain events detectable
The lack of success of rTMS in MD can be explained with the EEG, namely, closed-loop stimulation. Kraus
also by technical reasons, the most obvious being the and colleagues181 suggested that it is possible to modify
large parameter space for rTMS. Studies in the literature cortical excitability by delivering TMS pulses during
have used very different TMS settings in terms of stimu- event-related desynchronization in the beta band during
lation frequency and intensity, duration of trains, inter- a motor imagery task, whereas Zrenner and col-
train interval, number of sessions, and time between leagues182 demonstrated that plasticity in M1 is more
them. This has very likely contributed to the observed easily induced if TMS pulses are delivered at negative
heterogeneity of results and has complicated the issue of peaks of the μ sensorimotor rhythm, which represents a
devising effective rTMS protocols. Some biological fac- state of increased excitability of corticospinal neurones.
tors should also be considered. For instance, it may be Although not yet investigated, the possibility to fine tune
necessary to target particular brain areas to treat specific the induction of plasticity by increasing spatial or tempo-
symptoms rather than applying a one-design-fits-all ral specificity may open new and exciting scenarios in
approach (the most effective therapeutic rTMS para- the treatment of MD.
digms are summarized in Table 3). Additional points Apart from technical issues, the lack of rTMS success
pertain to the low number of participants included in as a therapeutic tool might lie in a more relevant prob-
many studies to possible interactions between drugs lem: our limited understanding of the mechanism of
and the effects of TMS and to an inappropriate dosage rTMS effects in humans. The first rTMS studies were
of TMS stimulation (ie, low number of pulses and/or based on a simple model in which low-frequency rTMS
session and low intensity).167 would weaken synapses and high-frequency rTMS
Another issue with rTMS protocols as a therapy is would strengthen synapses, thereby increasing or decreas-
that their effects are remarkably variable across subjects ing the excitability of the target area. It was based on
as well as within individuals.168-172 Part of this problem data from experiments carried out in tissue slices of
might be addressed by increasing the specificity of TMS the hippocampal formation from dead rodents, but
by changing the stimulation parameters. Novel TMS whether these mechanisms are also relevant in the neo-
devices, such as controllable TMS (cTMS),173 allow cortex of awake, behaving humans is not known. It is
changes in the duration and shape of magnetic stimuli. likely, in fact, that the sensitivity of different structures
By varying them, it is possible to activate different sub- to synaptic plasticity varies across the brain.183 Indeed,
sets of inputs to M1.174,175 This notion can be applied in awake, freely moving rats it is much more difficult to
to rTMS as well; indeed, it has been demonstrated that sustain changes in neocortical synaptic plasticity using
the effects of 1 Hz rTMS are greater when monophasic conventional repetitive stimulation protocols than in
stimuli delivered via a cTMS device are used when com- slice preparations.184 The limited evidence we have in
pared with standard biphasic pulses.176 There is also humans is that activation of NMDA receptors is impor-
preliminary evidence that the effects of TBS on M1 are tant in explaining some of the effects of rTMS in the
determined more by the pulse characteristics, and thus motor cortex.185,186 Thus, some changes at the synaptic
potentially by the specific neural population recruited, level seem to be involved in the after-effects of rTMS,
rather than the TBS pattern (intermittent or continu- but how these exactly map onto the various types and
ous).169 A second way to achieve greater specificity of stages of the LTP/LTD described in animals is still
TMS is to use the high time resolution of EEG to deliver unclear. Another complicating factor is that TMS

TABLE 3. Summary of the most effective therapeutic rTMS paradigms in each movement disorders condition

PD (motor symptoms) PD (depression) Dystonia TS HD ET

rTMS protocol High-frequency on M1 High-frequency Low-frequency Low-frequency Controversial Low-frequency


or low-frequency rTMS on left rTMS on dorsal rTMS on SMA data rTMS on
rTMS applied over DLPFC PMC cerebellum
other frontal regions

DLPFC, dorsolateral prefrontal cortex; ET, essential tremor; HD, Huntington’s disease; PD, Parkinson’s disease; PMC, premotor cortex; rTMS, repetitive trans-
cranial magnetic stimulation; SMA, supplementary motor area; TS, Tourette syndrome.

8 Movement Disorders, 2019


R T M S F O R T H E T R E A T M E N T O F M O V E M E N T D I S O R D E R S

activates a large number of presynaptic and postsynaptic perceptual improvement in healthy subjects. Clin Neurophysiol
2017;128(6):1015-1025.
cellular structures simultaneously, causing a massive syn-
13. Davare M, Montague K, Olivier E, Rothwell JC, Lemon RN. Ven-
aptic bombardment of excitatory and inhibitory cells.183 tral premotor to primary motor cortical interactions during object-
Thus, it is impossible to predict how individual neural driven grasp in humans. Cortex 2009;45(9):1050-1057.
types will respond to any protocol. More important, we 14. Suppa A, Rocchi L, Li Voti P, et al. The Photoparoxysmal
do not understand how the effects of rTMS at the site of Response Reflects Abnormal Early Visuomotor Integration in the
Human Motor Cortex. Brain Stim 2015;8(6):1151-1161.
stimulation translate into effects at a circuit level, which
15. Murase N, Cengiz B, Rothwell JC. Inter-individual variation in the
might be the critical issue in MD.187 All of these consid- after-effect of paired associative stimulation can be predicted from
erations seem to suggest that the term LTP/LTD-like short-interval intracortical inhibition with the threshold tracking
plasticity, commonly used to describe the rTMS-induced method. Brain Stim 2015;8(1):105-113.

effects, might not accurately reflect the underlying mech- 16. Suppa A, Biasiotta A, Belvisi D, et al. Heat-evoked experimental
pain induces long-term potentiation-like plasticity in human pri-
anisms that occur in the human brain. Unfortunately, mary motor cortex. Cereb Cortex 2013;23(8):1942-1951.
there are no physiological markers in humans specific 17. Berardelli A, Inghilleri M, Rothwell JC, et al. Facilitation of muscle
for synaptic plasticity, so further work is certainly evoked responses after repetitive cortical stimulation in man. Exp
required if rTMS is to be helpful in treating plasticity- Brain Res 1998;122(1):79-84.

related abnormalities in the human brain. 18. Conte A, Li Voti P, Pontecorvo S, et al. Attention-related changes
in short-term cortical plasticity help to explain fatigue in multiple
In conclusion, rTMS has a neuromodulatory poten- sclerosis. Mult Scler 2016;22(10):1359-1366.
tial that might be successfully used in clinical practice. 19. Huang YZ, Edwards MJ, Rounis E, Bhatia KP, Rothwell JC. Theta
However, a better understanding of the underlying burst stimulation of the human motor cortex. Neuron 2005;45(2):
mechanisms, together with an optimization of stimula- 201-206.

tion protocols, are needed for a more effective applica- 20. Fitzgerald PB, Fountain S, Daskalakis ZJ. A comprehensive review
of the effects of rTMS on motor cortical excitability and inhibition.
tion of rTMS as a therapy in MD. Clin Neurophysiol 2006;117(12):2584-2596.
21. Bologna M, Rocchi L, Paparella G, et al. Reversal of practice-
related effects on corticospinal excitability has no immediate effect
Acknowledgment: We confirm that we have read the journal’s position on behavioral outcome. Brain Stim 2015;8(3):603-612.
on issues involved in ethical publication and affirm that this work is con-
sistent with those guidelines. 22. Georgiev D, Rocchi L, Tocco P, Speekenbrink M, Rothwell JC,
Jahanshahi M. Continuous theta burst stimulation over the dorso-
lateral prefrontal cortex and the pre-SMA alter drift rate and
response thresholds respectively during perceptual decision-making.
Brain Stim 2016;9(4):601-608.
References
23. Nardella A, Rocchi L, Conte A, Bologna M, Suppa A, Berardelli A.
1. Barker AT, Jalinous R, Freeston IL. Non-invasive magnetic stimula- Inferior parietal lobule encodes visual temporal resolution pro-
tion of human motor cortex. Lancet 1985;1(8437):1106-1107. cesses contributing to the critical flicker frequency threshold in
2. Rothwell JC. Techniques and mechanisms of action of transcranial humans. PloS ONE 2014;9(6):e98948.
stimulation of the human motor cortex. J Neurosci Methods 1997; 24. Mendez JC, Rocchi L, Jahanshahi M, Rothwell J, Merchant H.
74(2):113-122. Probing the timing network: A continuous theta burst stimulation
3. Cantello R. Applications of transcranial magnetic stimulation in study of temporal categorization. Neuroscience 2017;356:167-175.
movement disorders. J Clin Neurophysiol 2002;19(4):272-293. 25. Casula EP, Pellicciari MC, Picazio S, Caltagirone C, Koch G.
4. Edwards MJ, Talelli P, Rothwell JC. Clinical applications of trans- Spike-timing-dependent plasticity in the human dorso-lateral pre-
cranial magnetic stimulation in patients with movement disorders. frontal cortex. NeuroImage 2016;143:204-213.
Lancet Neurol 2008;7(9):827-840. 26. Chen R, Cros D, Curra A, et al. The clinical diagnostic utility of
5. Barker AT. An introduction to the basic principles of magnetic transcranial magnetic stimulation: report of an IFCN committee.
nerve stimulation. J Clin Neurophysiol 1991;8(1):26-37. Clin Neurophysiol 2008;119(3):504-532.
6. Rothwell JC. Using transcranial magnetic stimulation methods to 27. Ziemann U, Paulus W, Nitsche MA, et al. Consensus: motor cortex
probe connectivity between motor areas of the brain. Hum Mov plasticity protocols. Brain Stim 2008;1(3):164-182.
Sci 2011;30(5):906-915. 28. Höflich G KS, Hufnagel S, Ruhrmann S, Möller HJ. Application of
7. Day BL, Dressler D, Maertens de Noordhout A, et al. Electric and transcranial magnetic stimulation in treatment of drug-resistant
magnetic stimulation of human motor cortex: surface EMG and major depression—a report of two cases. Hum Psychopharmacol
single motor unit responses. J Physiol 1989;412:449-473. 1993;8:361-365.
8. Nakamura H, Kitagawa H, Kawaguchi Y, Tsuji H. Intracortical 29. Di Lazzaro V, Rothwell J, Capogna M. Noninvasive stimulation of
facilitation and inhibition after transcranial magnetic stimulation in the human brain: activation of multiple cortical circuits. Neurosci-
conscious humans. J Physiol 1997;498 (Pt 3):817-823. entist 2018;24(3):246-260.
9. Valls-Sole J, Pascual-Leone A, Wassermann EM, Hallett M. Human 30. Bliss TV, Cooke SF. Long-term potentiation and long-term depres-
motor evoked responses to paired transcranial magnetic stimuli. sion: a clinical perspective. Clinics (Sao Paulo) 2011;66(suppl 1):3-17.
Electroencephalogr Clin Neurophysiol 1992;85(6):355-364. 31. Hoogendam JM, Ramakers GM, Di Lazzaro V. Physiology of
10. Kujirai T, Caramia MD, Rothwell JC, et al. Corticocortical inhibi- repetitive transcranial magnetic stimulation of the human brain.
tion in human motor cortex. J Physiol 1993;471:501-519. Brain Stim 2010;3(2):95-118.
11. Ziemann U, Tergau F, Wassermann EM, Wischer S, Hildebrandt J, 32. Cirillo G, Di Pino G, Capone F, et al. Neurobiological after-effects
Paulus W. Demonstration of facilitatory I wave interaction in the of non-invasive brain stimulation. Brain Stim 2017;10(1):1-18.
human motor cortex by paired transcranial magnetic stimulation.
33. Gersner R, Kravetz E, Feil J, Pell G, Zangen A. Long-term effects
J Physiol 1998;511(Pt 1):181-190.
of repetitive transcranial magnetic stimulation on markers for neu-
12. Rocchi L, Erro R, Antelmi E, et al. High frequency somatosensory roplasticity: differential outcomes in anesthetized and awake ani-
stimulation increases sensori-motor inhibition and leads to mals. J Neurosci 2011;31(20):7521-7526.

Movement Disorders, 2019 9


L A T O R R E E T A L

34. Mix A, Benali A, Funke K. Strain differences in the effect of rTMS 54. Cummings JL. The neuroanatomy of depression. J Clin Psychiatry
on cortical expression of calcium-binding proteins in rats. Exp 1993;54(suppl):14-20.
Brain Res 2014;232(2):435-442. 55. Kito S, Fujita K, Koga Y. Changes in regional cerebral blood flow
35. Stefan K, Kunesch E, Cohen LG, Benecke R, Classen J. Induction after repetitive transcranial magnetic stimulation of the left dorso-
of plasticity in the human motor cortex by paired associative stimu- lateral prefrontal cortex in treatment-resistant depression.
lation. Brain 2000;123 Pt 3:572-584. J Neuropsychiatry Clin Neurosci 2008;20(1):74-80.
36. Hedges DW, Salyer DL, Higginbotham BJ, et al. Transcranial mag- 56. Paus T, Barrett J. Transcranial magnetic stimulation (TMS) of the
netic stimulation (TMS) effects on testosterone, prolactin, and cor- human frontal cortex: implications for repetitive TMS treatment of
ticosterone in adult male rats. Biol Psychiatry 2002;51(5):417-421. depression. J Psychiatry Neurosci 2004;29(4):268-279.
37. Baeken C, Vanderhasselt MA, Remue J, et al. One left dorsolateral 57. Hamilton JP, Chen MC, Gotlib IH. Neural systems approaches to
prefrontal cortical HF-rTMS session attenuates HPA-system sensi- understanding major depressive disorder: an intrinsic functional
tivity to critical feedback in healthy females. Neuropsychologia organization perspective. Neurobiol Dis 2013;52:4-11.
2014;57:112-121. 58. Dunlop K, Hanlon CA, Downar J. Noninvasive brain stimulation
38. Grehl S, Viola HM, Fuller-Carter PI, et al. Cellular and molecular treatments for addiction and major depression. Ann N Y Acad Sci
changes to cortical neurons following low intensity repetitive mag- 2017;1394(1):31-54.
netic stimulation at different frequencies. Brain Stim 2015;8(1): 59. Koenigs M, Grafman J. The functional neuroanatomy of depres-
114-123. sion: distinct roles for ventromedial and dorsolateral prefrontal
cortex. Behav Brain Res 2009;201(2):239-243.
39. Ji RR, Schlaepfer TE, Aizenman CD, et al. Repetitive transcranial
magnetic stimulation activates specific regions in rat brain. Proc 60. Mayberg HS, Lozano AM, Voon V, et al. Deep brain stimulation
Natl Acad Sci U S A 1998;95(26):15635-15640. for treatment-resistant depression. Neuron 2005;45(5):651-660.
40. Abraham WC. How long will long-term potentiation last? Philos 61. Brody AL, Saxena S, Mandelkern MA, Fairbanks LA, Ho ML,
Trans R Soc Lond B Biol Sci 2003;358(1432):735-744. Baxter LR. Brain metabolic changes associated with symptom fac-
tor improvement in major depressive disorder. Biol Psychiatry
41. Baumer T, Demiralay C, Hidding U, et al. Abnormal plasticity of 2001;50(3):171-178.
the sensorimotor cortex to slow repetitive transcranial magnetic
stimulation in patients with writer’s cramp. Mov Disord 2007;22 62. Ge R, Blumberger DM, Downar J, et al. Abnormal functional con-
(1):81-90. nectivity within resting-state networks is related to rTMS-based
therapy effects of treatment resistant depression: a pilot study.
42. Khedr EM, Kotb H, Kamel NF, Ahmed MA, Sadek R, Rothwell JC. J Affect Disord 2017;218:75-81.
Longlasting antalgic effects of daily sessions of repetitive transcranial
magnetic stimulation in central and peripheral neuropathic pain. 63. Shajahan PM, Glabus MF, Steele JD, et al. Left dorso-lateral repeti-
J Neurol Neurosurg Psychiatry 2005;76(6):833-838. tive transcranial magnetic stimulation affects cortical excitability
and functional connectivity, but does not impair cognition in major
43. Borich M, Arora S, Kimberley TJ. Lasting effects of repeated rTMS depression. Prog Neuropsychopharmacol Biol Psychiatry 2002;26
application in focal hand dystonia. Restor Neurol Neurosci 2009; (5):945-954.
27(1):55-65.
64. Loo CK, Sachdev PS, Haindl W, et al. High (15 Hz) and low
44. Schulze L, Feffer K, Lozano C, et al. Number of pulses or number (1 Hz) frequency transcranial magnetic stimulation have different
of sessions? An open-label study of trajectories of improvement for acute effects on regional cerebral blood flow in depressed patients.
once-vs. twice-daily dorsomedial prefrontal rTMS in major depres- Psychol Med 2003;33(6):997-1006.
sion. Brain Stim 2018;11(2):327-336. 65. Michael N, Gosling M, Reutemann M, et al. Metabolic changes
45. Lefaucheur JP, Andre-Obadia N, Antal A, et al. Evidence-based after repetitive transcranial magnetic stimulation (rTMS) of the left
guidelines on the therapeutic use of repetitive transcranial magnetic prefrontal cortex: a sham-controlled proton magnetic resonance
stimulation (rTMS). Clin Neurophysiol 2014;125(11):2150-2206. spectroscopy (1H MRS) study of healthy brain. Eur J Neurosci
2003;17(11):2462-2468.
46. Milev RV, Giacobbe P, Kennedy SH, et al. Canadian Network for
Mood and Anxiety Treatments (CANMAT) 2016 clinical guide- 66. Pogarell O, Koch W, Popperl G, et al. Striatal dopamine release
lines for the management of adults with major depressive disorder: after prefrontal repetitive transcranial magnetic stimulation in
section 4. neurostimulation treatments. Can J Psychiatry 2016;61(9): major depression: preliminary results of a dynamic [123I] IBZM
561-575. SPECT study. J Psychiatr Res 2006;40(4):307-314.

47. O’Reardon JP, Solvason HB, Janicak PG, et al. Efficacy and safety 67. Luborzewski A, Schubert F, Seifert F, et al. Metabolic alterations in
of transcranial magnetic stimulation in the acute treatment of the dorsolateral prefrontal cortex after treatment with high-frequency
major depression: a multisite randomized controlled trial. Biol Psy- repetitive transcranial magnetic stimulation in patients with unipolar
chiatry 2007;62(11):1208-1216. major depression. J Psychiatr Res 2007;41(7):606-615.
68. Zwanzger P, Baghai TC, Padberg F, et al. The combined
48. McClintock SM, Reti IM, Carpenter LL, et al. Consensus Recom-
dexamethasone-CRH test before and after repetitive transcranial
mendations for the Clinical Application of Repetitive Transcranial
magnetic stimulation (rTMS) in major depression. Psychoneuro-
Magnetic Stimulation (rTMS) in the treatment of depression. J Clin
endocrinology 2003;28(3):376-385.
Psychiatry 2018;79(1):35-48.
69. Yukimasa T, Yoshimura R, Tamagawa A, et al. High-frequency
49. Yip AG, George MS, Tendler A, Roth Y, Zangen A, Carpenter LL. repetitive transcranial magnetic stimulation improves refractory
61% of unmedicated treatment resistant depression patients who depression by influencing catecholamine and brain-derived neuro-
did not respond to acute TMS treatment responded after four trophic factors. Pharmacopsychiatry 2006;39(2):52-59.
weeks of twice weekly deep TMS in the Brainsway pivotal trial.
Brain Stim 2017;10(4):847-849. 70. Zanardini R, Gazzoli A, Ventriglia M, et al. Effect of repetitive
transcranial magnetic stimulation on serum brain derived neuro-
50. Anderson RJ, Hoy KE, Daskalakis ZJ, Fitzgerald PB. Repetitive trophic factor in drug resistant depressed patients. J Affect Disord
transcranial magnetic stimulation for treatment resistant depres- 2006;91(1):83-86.
sion: re-establishing connections. Clin Neurophysiol 2016;127(11):
3394-3405. 71. Herwig U, Fallgatter AJ, Hoppner J, et al. Antidepressant effects of
augmentative transcranial magnetic stimulation: randomised multi-
51. Bolognini N, Pascual-Leone A, Fregni F. Using non-invasive brain centre trial. Br J Psychiatry 2007;191:441-448.
stimulation to augment motor training-induced plasticity. J Neuroeng
Rehabil 2009;6:8. 72. George MS, Lisanby SH, Avery D, et al. Daily left prefrontal trans-
cranial magnetic stimulation therapy for major depressive disorder:
52. Rossi S, Rossini PM. TMS in cognitive plasticity and the potential a sham-controlled randomized trial. Arch Gen Psychiatry 2010;67(5):
for rehabilitation. Trends Cogn Sci 2004;8(6):273-279. 507-516.
53. Talelli P, Rothwell J. Does brain stimulation after stroke have a 73. Yesavage JA, Fairchild JK, Mi Z, et al. Effect of repetitive trans-
future? Curr Opin Neurol 2006;19(6):543-550. cranial magnetic stimulation on treatment-resistant major

10 Movement Disorders, 2019


R T M S F O R T H E T R E A T M E N T O F M O V E M E N T D I S O R D E R S

depression in US veterans: a randomized clinical trial. JAMA Psy- 92. Little S, Brown P. The functional role of beta oscillations in
chiatry 2018;75(9):884-893. Parkinson’s disease. Parkinsonism Relat Disord 2014;20(suppl 1):
74. Carpenter LL, Janicak PG, Aaronson ST, et al. Transcranial mag- S44-S48.
netic stimulation (TMS) for major depression: a multisite, naturalis- 93. Sanders TH, Jaeger D. Optogenetic stimulation of cortico-
tic, observational study of acute treatment outcomes in clinical subthalamic projections is sufficient to ameliorate bradykinesia in
practice. Depress Anxiety 2012;29(7):587-596. 6-ohda lesioned mice. Neurobiol Dis 2016;95:225-237.
75. Mantovani A, Pavlicova M, Avery D, et al. Long-term efficacy of 94. Drouot X, Oshino S, Jarraya B, et al. Functional recovery in a pri-
repeated daily prefrontal transcranial magnetic stimulation (TMS) mate model of Parkinson’s disease following motor cortex stimula-
in treatment-resistant depression. Depress Anxiety 2012;29(10): tion. Neuron 2004;44(5):769-778.
883-890. 95. Moro E, Schwalb JM, Piboolnurak P, et al. Unilateral subdural
76. McDonald WM, Durkalski V, Ball ER, et al. Improving the antide- motor cortex stimulation improves essential tremor but not
pressant efficacy of transcranial magnetic stimulation: maximizing Parkinson’s disease. Brain 2011;134(Pt 7):2096-2105.
the number of stimulations and treatment location in treatment-
96. Siebner HR, Rossmeier C, Mentschel C, Peinemann A, Conrad B.
resistant depression. Depress Anxiety 2011;28(11):973-980.
Short-term motor improvement after sub-threshold 5-Hz repetitive
77. Connolly KR, Helmer A, Cristancho MA, Cristancho P, O’Reardon transcranial magnetic stimulation of the primary motor hand area
JP. Effectiveness of transcranial magnetic stimulation in clinical prac- in Parkinson’s disease. J Neurol Sci 2000;178(2):91-94.
tice post-FDA approval in the United States: results observed with
97. Shimamoto H, Takasaki K, Shigemori M, Imaizumi T, Ayabe M,
the first 100 consecutive cases of depression at an academic medical
Shoji H. Therapeutic effect and mechanism of repetitive trans-
center. J Clin Psychiatry 2012;73(4):e567-573.
cranial magnetic stimulation in Parkinson’s disease. J Neurol 2001;
78. Berlim MT, Van den Eynde F, Daskalakis ZJ. A systematic review 248(suppl 3):III48-III52.
and meta-analysis on the efficacy and acceptability of bilateral 98. Khedr EM, Farweez HM, Islam H. Therapeutic effect of repetitive
repetitive transcranial magnetic stimulation (rTMS) for treating
transcranial magnetic stimulation on motor function in Parkinson’s
major depression. Psychol Med 2013;43(11):2245-2254.
disease patients. Eur J Neurol 2003;10(5):567-572.
79. Berlim MT, Van den Eynde F, Daskalakis ZJ. Efficacy and accept- 99. Ikeguchi M, Touge T, Nishiyama Y, Takeuchi H, Kuriyama S,
ability of high frequency repetitive transcranial magnetic stimula- Ohkawa M. Effects of successive repetitive transcranial magnetic
tion (rTMS) versus electroconvulsive therapy (ECT) for major stimulation on motor performances and brain perfusion in idio-
depression: a systematic review and meta-analysis of randomized pathic Parkinson’s disease. J Neurol Sci 2003;209(1-2):41-46.
trials. Depress Anxiety 2013;30(7):614-623.
100. Okabe S, Ugawa Y, Kanazawa I, Effectiveness of rTMS on
80. Berlim MT, Van den Eynde F, Daskalakis ZJ. High-frequency Parkinson’s Disease Study Group. 0.2-Hz repetitive transcranial
repetitive transcranial magnetic stimulation accelerates and magnetic stimulation has no add-on effects as compared to a realis-
enhances the clinical response to antidepressants in major depres- tic sham stimulation in Parkinson’s disease. Mov Disord 2003;18
sion: a meta-analysis of randomized, double-blind, and sham- (4):382-388.
controlled trials. J Clin Psychiatry 2013;74(2):e122-129.
101. Lefaucheur JP, Drouot X, Von Raison F, Menard-Lefaucheur I,
81. Berlim MT, Van den Eynde F, Jeff Daskalakis Z. Clinically mean- Cesaro P, Nguyen JP. Improvement of motor performance and
ingful efficacy and acceptability of low-frequency repetitive trans- modulation of cortical excitability by repetitive transcranial mag-
cranial magnetic stimulation (rTMS) for treating primary major netic stimulation of the motor cortex in Parkinson’s disease. Clin
depression: a meta-analysis of randomized, double-blind and sham- Neurophysiol 2004;115(11):2530-2541.
controlled trials. Neuropsychopharmacology 2013;38(4):543-551.
102. Bornke C, Schulte T, Przuntek H, Muller T. Clinical effects of
82. Schutter DJ. Quantitative review of the efficacy of slow-frequency repetitive transcranial magnetic stimulation versus acute levodopa
magnetic brain stimulation in major depressive disorder. Psychol challenge in Parkinson’s disease. J Neural Transm Supp 2004(68):
Med 2010;40(11):1789-1795. 61-67.
83. Gaynes BN, Lux LJ, Lloyd SW, et al. Nonpharmacologic Interven- 103. Hamada M, Ugawa Y, Tsuji S, Effectiveness of rTMS on Parkinson’s
tions for Treatment-Resistant Depression in Adults. Rockville, Disease Study Group. High-frequency rTMS over the supplementary
MD: Agency for Healthcare Research and Quality (US); (Compara- motor area for treatment of Parkinson’s disease. Mov Disord 2008;
tive Effectiveness Reviews, No. 33.) 2011. 23(11):1524-1531.
84. George MS, Schlaepfer T, Padberg F, Fitzgerald PB. Brain stimula- 104. Sedlackova S, Rektorova I, Srovnalova H, Rektor I. Effect of high
tion treatments for depression. World J Biol Psychiatry 2014;15(2): frequency repetitive transcranial magnetic stimulation on reaction
167-168. time, clinical features and cognitive functions in patients with
85. Kennedy SH, Milev R, Giacobbe P, et al. Canadian Network for Parkinson’s disease. J Neural Transm 2009;116(9):1093-1101.
Mood and Anxiety Treatments (CANMAT) clinical guidelines for 105. Arias P, Vivas J, Grieve KL, Cudeiro J. Controlled trial on the
the management of major depressive disorder in adults. effect of 10 days low-frequency repetitive transcranial magnetic
IV. Neurostimulation therapies. J Affect Disord 2009;117(suppl 1): stimulation (rTMS) on motor signs in Parkinson’s disease. Mov
S44-S53. Disord 2010;25(12):1830-1838.
86. DeLong MR, Wichmann T. Basal ganglia circuits as targets for 106. Kimura H, Kurimura M, Kurokawa K, et al. A comprehensive
neuromodulation in Parkinson disease. JAMA Neurol 2015;72 study of repetitive transcranial magnetic stimulation in Parkinson’s
(11):1354-1360. disease. ISRN Neurol 2011;2011:845453.
87. Watanabe T, Hanajima R, Shirota Y, et al. Effects of rTMS of pre- 107. Benninger DH, Iseki K, Kranick S, Luckenbaugh DA, Houdayer E,
supplementary motor area on fronto basal ganglia network activity Hallett M. Controlled study of 50-Hz repetitive transcranial mag-
during stop-signal task. J Neurosci 2015;35(12):4813-4823. netic stimulation for the treatment of Parkinson disease.
88. Bestmann S, Baudewig J, Siebner HR, Rothwell JC, Frahm J. Neurorehabil Neural Repair 2012;26(9):1096-1105.
BOLD MRI responses to repetitive TMS over human dorsal 108. Shirota Y, Ohtsu H, Hamada M, Enomoto H, Ugawa Y, Research
premotor cortex. NeuroImage 2005;28(1):22-29. Committee on r TMS treatment of Parkinson’s Disease
89. Strafella AP, Paus T, Fraraccio M, Dagher A. Striatal dopamine (TMSToPsD). Supplementary motor area stimulation for Parkinson
release induced by repetitive transcranial magnetic stimulation of disease: a randomized controlled study. Neurology 2013;80(15):
the human motor cortex. Brain 2003;126(Pt 12):2609-2615. 1400-1405.
90. Kim JY, Chung EJ, Lee WY, et al. Therapeutic effect of repetitive 109. Maruo T, Hosomi K, Shimokawa T, et al. High-frequency repeti-
transcranial magnetic stimulation in Parkinson’s disease: analysis tive transcranial magnetic stimulation over the primary foot motor
of [11C] raclopride PET study. Mov Disord 2008;23(2):207-211. area in Parkinson’s disease. Brain Stim 2013;6(6):884-891.
91. Gaynor LM, Kuhn AA, Dileone M, et al. Suppression of beta oscil- 110. Brys M, Fox MD, Agarwal S, et al. Multifocal repetitive TMS for
lations in the subthalamic nucleus following cortical stimulation in motor and mood symptoms of Parkinson disease: a randomized
humans. Eur J Neurosci 2008;28(8):1686-1695. trial. Neurology 2016;87(18):1907-1915.

Movement Disorders, 2019 11


L A T O R R E E T A L

111. Mally J, Geisz N, Dinya E. Follow up study: the influence of rTMS 131. Xie CL, Chen J, Wang XD, et al. Repetitive transcranial magnetic
with high and low frequency stimulation on motor and executive stimulation (rTMS) for the treatment of depression in Parkinson
function in Parkinson’s disease. Brain Res Bull 2017;135:98-104. disease: a meta-analysis of randomized controlled clinical trials.
112. Yokoe M, Mano T, Maruo T, et al. The optimal stimulation site Neurol Sci 2015;36(10):1751-1761.
for high-frequency repetitive transcranial magnetic stimulation in 132. Fregni F, Santos CM, Myczkowski ML, et al. Repetitive trans-
Parkinson’s disease: a double-blind crossover pilot study. J Clin cranial magnetic stimulation is as effective as fluoxetine in the
Neurosci 2018;47:72-78. treatment of depression in patients with Parkinson’s disease.
113. Filipovic SR, Rothwell JC, Bhatia K. Low-frequency repetitive J Neurol Neurosurg Psychiatry 2004;75(8):1171-1174.
transcranial magnetic stimulation and off-phase motor symptoms 133. Benninger DH, Berman BD, Houdayer E, et al. Intermittent theta-
in Parkinson’s disease. J Neurol Sci 2010;291(1-2):1-4. burst transcranial magnetic stimulation for treatment of Parkinson
114. Khedr EM, Rothwell JC, Shawky OA, Ahmed MA, Foly N, disease. Neurology 2011;76(7):601-609.
Hamdy A. Dopamine levels after repetitive transcranial magnetic 134. Boggio PS, Fregni F, Bermpohl F, et al. Effect of repetitive TMS
stimulation of motor cortex in patients with Parkinson’s disease: and fluoxetine on cognitive function in patients with Parkinson’s
preliminary results. Mov Disord 2007;22(7):1046-1050. disease and concurrent depression. Mov Disord 2005;20(9):1178-
115. Boylan LS, Pullman SL, Lisanby SH, Spicknall KE, Sackeim HA. 1184.
Repetitive transcranial magnetic stimulation to SMA worsens com- 135. Cardoso EF, Fregni F, Martins Maia F, et al. rTMS treatment for
plex movements in Parkinson’s disease. Clin Neurophysiol 2001; depression in Parkinson’s disease increases BOLD responses in the
112(2):259-264. left prefrontal cortex. Int J Neuropsychopharmacol 2008;11(2):
116. Rothkegel H, Sommer M, Rammsayer T, Trenkwalder C, 173-183.
Paulus W. Training effects outweigh effects of single-session con- 136. Pal E, Nagy F, Aschermann Z, Balazs E, Kovacs N. The impact of
ventional rTMS and theta burst stimulation in PD patients. left prefrontal repetitive transcranial magnetic stimulation on
Neurorehabil Neural Repair 2009;23(4):373-381. depression in Parkinson’s disease: a randomized, double-blind,
117. Acosta-Cabronero J, Cardenas-Blanco A, Betts MJ, et al. The placebo-controlled study. Mov Disord 2010;25(14):2311-2317.
whole-brain pattern of magnetic susceptibility perturbations in 137. Makkos A, Pal E, Aschermann Z, et al. High-frequency repetitive
Parkinson’s disease. Brain 2017;140(1):118-131. transcranial magnetic stimulation can improve depression in
118. Khedr EM, Rothwell JC, Shawky OA, Ahmed MA, Hamdy A. Parkinson’s disease: a randomized, double-blind, placebo-
Effect of daily repetitive transcranial magnetic stimulation on motor controlled study. Neuropsychobiology 2016;73(3):169-177.
performance in Parkinson’s disease. Mov Disord 2006;21(12):2201- 138. Shin HW, Youn YC, Chung SJ, Sohn YH. Effect of high-frequency
2205. repetitive transcranial magnetic stimulation on major depressive
119. Chou YH, Hickey PT, Sundman M, Song AW, Chen NK. Effects disorder in patients with Parkinson’s disease. J Neurol 2016;263
of repetitive transcranial magnetic stimulation on motor symptoms (7):1442-1448.
in Parkinson disease: a systematic review and meta-analysis. JAMA 139. Quartarone A, Hallett M. Emerging concepts in the physiological
Neurol 2015;72(4):432-440. basis of dystonia. Mov Disord 2013;28(7):958-967.
120. Zanjani A, Zakzanis KK, Daskalakis ZJ, Chen R. Repetitive trans-
140. Erro R, Tinazzi M, Morgante F, Bhatia KP. Non-invasive brain
cranial magnetic stimulation of the primary motor cortex in the
stimulation for dystonia: therapeutic implications. Eur J Neurol
treatment of motor signs in Parkinson’s disease: a quantitative
2017;24(10):1228-e1264.
review of the literature. Mov Disord 2015;30(6):750-758.
141. Bradnam LV, McDonnell MN, Ridding MC. Cerebellar intermit-
121. Wagle-Shukla A, Angel MJ, Zadikoff C, et al. Low-frequency
tent theta-burst stimulation and motor control training in individ-
repetitive transcranial magnetic stimulation for treatment of
uals with cervical dystonia. Brain Sci 2016;6(4):56.
levodopa-induced dyskinesias. Neurology 2007;68(9):704-705.
122. Filipovic SR, Rothwell JC, van de Warrenburg BP, Bhatia K. 142. Huang YZ, Rothwell JC, Lu CS, Wang J, Chen RS. Restoration of
Repetitive transcranial magnetic stimulation for levodopa-induced motor inhibition through an abnormal premotor-motor connection
dyskinesias in Parkinson’s disease. Mov Disord 2009;24(2): in dystonia. Mov Disord 2010;25(6):696-703.
246-253. 143. Cunnington R, Windischberger C, Moser E. Premovement activity
123. Koch G, Brusa L, Caltagirone C, et al. rTMS of supplementary of the pre-supplementary motor area and the readiness for action:
motor area modulates therapy-induced dyskinesias in Parkinson studies of time-resolved event-related functional MRI. Hum Mov
disease. Neurology 2005;65(4):623-625. Sci 2005;24(5-6):644-656.

124. Koch G, Brusa L, Carrillo F, et al. Cerebellar magnetic stimulation 144. Leckman JF BM, Sukhodolsky DG. Phenomenology of tics and
decreases levodopa-induced dyskinesias in Parkinson disease. Neu- sensory urges: the self under siege. In: Martino D LJ, ed. Tourette
rology 2009;73(2):113-119. Syndrome. New York: Oxford University Press, 2013:3-25.
125. Sayin S, Cakmur R, Yener GG, Yaka E, Ugurel B, Uzunel F. Low- 145. Bloch MH, Peterson BS, Scahill L, et al. Adulthood outcome of tic
frequency repetitive transcranial magnetic stimulation for dyskine- and obsessive-compulsive symptom severity in children with
sia and motor performance in Parkinson’s disease. J Clin Neurosci Tourette syndrome. Arch Pediatr Adolesc Med 2006;160(1):65-69.
2014;21(8):1373-1376. 146. Bohlhalter S, Goldfine A, Matteson S, et al. Neural correlates of tic
126. Cerasa A, Koch G, Donzuso G, et al. A network centred on the generation in Tourette syndrome: an event-related functional MRI
inferior frontal cortex is critically involved in levodopa-induced study. Brain 2006;129(Pt 8):2029-2037.
dyskinesias. Brain 2015;138(Pt 2):414-427. 147. Mantovani A, Lisanby SH, Pieraccini F, Ulivelli M, Castrogiovanni P,
127. Kim MS, Chang WH, Cho JW, et al. Efficacy of cumulative high- Rossi S. Repetitive transcranial magnetic stimulation (rTMS) in the
frequency rTMS on freezing of gait in Parkinson’s disease. Restor treatment of obsessive-compulsive disorder (OCD) and Tourette’s syn-
Neurol Neurosci 2015;33(4):521-530. drome (TS). Int J Neuropsychopharmacol 2006;9(1):95-100.
128. Kim SJ, Paeng SH, Kang SY. Stimulation in supplementary motor 148. Kwon HJ, Lim WS, Lim MH, et al. 1-Hz low frequency repetitive
area versus motor cortex for freezing of gait in Parkinson’s disease. transcranial magnetic stimulation in children with Tourette’s syn-
J Clin Neurol 2018;14(3):320-326. drome. Neurosci Lett 2011;492(1):1-4.
129. Tard C, Devanne H, Defebvre L, Delval A. Single session intermit- 149. Le K, Liu L, Sun M, Hu L, Xiao N. Transcranial magnetic stimula-
tent theta-burst stimulation on the left premotor cortex does not tion at 1 Hertz improves clinical symptoms in children with Tourette
alleviate freezing of gait in Parkinson’s disease. Neurosci Lett syndrome for at least 6 months. J Clin Neurosci 2013;20(2):
2016;628:1-9. 257-262.
130. Bologna M, Di Biasio F, Conte A, Iezzi E, Modugno N, 150. Landeros-Weisenberger A, Mantovani A, Motlagh MG, et al. Ran-
Berardelli A. Effects of cerebellar continuous theta burst stimula- domized sham controlled double-blind trial of repetitive trans-
tion on resting tremor in Parkinson’s disease. Parkinsonism Relat cranial magnetic stimulation for adults with severe Tourette
Disord 2015;21(9):1061-1066. syndrome. Brain Stim 2015;8(3):574-581.

12 Movement Disorders, 2019


R T M S F O R T H E T R E A T M E N T O F M O V E M E N T D I S O R D E R S

151. Wu SW, Maloney T, Gilbert DL, et al. Functional MRI-navigated 172. Lopez-Alonso V, Cheeran B, Rio-Rodriguez D, Fernandez-Del-
repetitive transcranial magnetic stimulation over supplementary Olmo M. Inter-individual variability in response to non-invasive
motor area in chronic tic disorders. Brain Stim 2014;7(2):212-218. brain stimulation paradigms. Brain Stim 2014;7(3):372-380.
152. Brusa L, Versace V, Koch G, et al. Improvement of choreic move- 173. Peterchev AV, D’Ostilio K, Rothwell JC, Murphy DL. Controllable
ments by 1 Hz repetitive transcranial magnetic stimulation in pulse parameter transcranial magnetic stimulator with enhanced cir-
Huntington’s disease patients. Ann Neurol 2005;58(4):655-656. cuit topology and pulse shaping J Neural Eng 2014;11(5):056023.
153. Shukla A, Jayarajan RN, Muralidharan K, Jain S. Repetitive trans- 174. D’Ostilio K, Goetz SM, Hannah R, et al. Effect of coil orientation
cranial magnetic stimulation not beneficial in severe choreiform on strength-duration time constant and I-wave activation with con-
movements of Huntington disease. J ECT 2013;29(2):e16-e17. trollable pulse parameter transcranial magnetic stimulation. Clin
Neurophysiol 2016;127(1):675-683.
154. Kloppel S, Draganski B, Siebner HR, Tabrizi SJ, Weiller C,
Frackowiak RS. Functional compensation of motor function in 175. Casula EP, Rocchi L, Hannah R, Rothwell JC. Effects of pulse
pre-symptomatic Huntington’s disease. Brain 2009;132(Pt 6):1624- width, waveform and current direction in the cortex: a combined
1632. cTMS-EEG study. Brain Stim 2018;11(5):1063-1070.
155. Groiss SJ, Netz J, Lange HW, Buetefisch CM. Frequency dependent 176. Goetz SM, Luber B, Lisanby SH, et al. Enhancement of
effects of rTMS on motor and cognitive functions in Huntington’s neuromodulation with novel pulse shapes generated by controllable
disease. Basal Ganglia 2012;2(1):41-48. pulse parameter transcranial magnetic stimulation. Brain Stim
2016;9(1):39-47.
156. Di Lazzaro V, Dileone M, Pilato F, et al. Repetitive transcranial
magnetic stimulation of the motor cortex for hemichorea. J Neurol 177. Keil J, Timm J, Sanmiguel I, Schulz H, Obleser J, Schonwiesner M.
Neurosurg Psychiatry 2006;77(9):1095-1097. Cortical brain states and corticospinal synchronization influence
TMS-evoked motor potentials. J Neurophysiol 2014;111(3):513-519.
157. Papoutsi M, Weiskopf N, Langbehn D, Reilmann R, Rees G,
Tabrizi SJ. Stimulating neural plasticity with real-time fMRI 178. Sauseng P, Klimesch W, Gerloff C, Hummel FC. Spontaneous
neurofeedback in Huntington’s disease: a proof of concept study. locally restricted EEG alpha activity determines cortical excitability
Hum Brain Mapp 2018;39(3):1339-1353. in the motor cortex. Neuropsychologia 2009;47(1):284-288.

158. Helmich RC, Toni I, Deuschl G, Bloem BR. The pathophysiology 179. Maki H, Ilmoniemi RJ. EEG oscillations and magnetically evoked
of essential tremor and Parkinson’s tremor. Curr Neurol Neurosci motor potentials reflect motor system excitability in overlapping
Rep 2013;13(9):378. neuronal populations. Clin Neurophysiol 2010;121(4):492-501.

159. Hallett M. Tremor: pathophysiology. Parkinsonism Relat Disord 180. Schulz H, Ubelacker T, Keil J, Muller N, Weisz N. Now I am
2014;20(suppl 1):S118-S122. ready-now i am not: the influence of pre-TMS oscillations and
corticomuscular coherence on motor-evoked potentials. Cereb Cor-
160. Gironell A, Kulisevsky J, Lorenzo J, Barbanoj M, Pascual- tex 2014;24(7):1708-1719.
Sedano B, Otermin P. Transcranial magnetic stimulation of the cer-
181. Kraus D, Naros G, Bauer R, et al. Brain state-dependent trans-
ebellum in essential tremor: a controlled study. Arch Neurol 2002;
cranial magnetic closed-loop stimulation controlled by sensorimo-
59(3):413-417.
tor desynchronization induces robust increase of corticospinal
161. Popa T, Russo M, Vidailhet M, et al. Cerebellar rTMS stimulation excitability. Brain Stim 2016;9(3):415-424.
may induce prolonged clinical benefits in essential tremor, and sub-
182. Zrenner C, Desideri D, Belardinelli P, Ziemann U. Real-time EEG-
jacent changes in functional connectivity: an open label trial. Brain
defined excitability states determine efficacy of TMS-induced plas-
Stim 2013;6(2):175-179.
ticity in human motor cortex. Brain Stim 2017;11(2):374-389.
162. Bologna M, Rocchi L, Leodori G, et al. Cerebellar continuous theta
183. Pell GS, Roth Y, Zangen A. Modulation of cortical excitability
burst stimulation in essential tremor. Cerebellum 2015;14(2):
induced by repetitive transcranial magnetic stimulation: influence
133-141.
of timing and geometrical parameters and underlying mechanisms.
163. Hellriegel H, Schulz EM, Siebner HR, Deuschl G, Raethjen JH. Progress Neurobiol 2011;93(1):59-98.
Continuous theta-burst stimulation of the primary motor cortex in 184. Boyd TE, Trepel C, Racine RJ. Cholinergic modulation of neocorti-
essential tremor. Clin Neurophysiol 2012;123(5):1010-1015. cal long-term potentiation in the awake, freely moving rat. Brain
164. Badran BW, Glusman CE, Austelle CW, et al. A double-blind, Res 2000;881(1):28-36.
sham-controlled pilot trial of pre-supplementary motor area (Pre- 185. Wankerl K, Weise D, Gentner R, Rumpf JJ, Classen J. L-type voltage-
SMA) 1 Hz rTMS to treat essential tremor. Brain Stim 2016;9(6): gated Ca2+ channels: a single molecular switch for long-term
945-947. potentiation/long-term depression-like plasticity and activity-dependent
165. Cagnan H, Brittain JS, Little S, et al. Phase dependent modulation metaplasticity in humans. J Neurosci 2010;30(18):6197-6204.
of tremor amplitude in essential tremor through thalamic stimula- 186. Huang YZ, Rothwell JC, Edwards MJ, Chen RS. Effect of physio-
tion. Brain 2013;136(Pt 10):3062-3075. logical activity on an NMDA-dependent form of cortical plasticity
166. Cagnan H, Pedrosa D, Little S, et al. Stimulating at the right time: in human. Cereb Cortex 2008;18(3):563-570.
phase-specific deep brain stimulation. Brain 2017;140(1):132-145. 187. Tang A, Thickbroom G, Rodger J. Repetitive transcranial magnetic
167. Chung CL, Mak MK. Effect of repetitive transcranial magnetic stimulation of the brain: mechanisms from animal and experimen-
stimulation on physical function and motor signs in Parkinson’s tal models. Neuroscientist 2015;23(1):82-94.
disease: a systematic review and meta-analysis. Brain Stim 2016;9 188. Pascual-Leone A, Valls-Sole J, Brasil-Neto JP, Cammarota A,
(4):475-487. Grafman J, Hallett M. Akinesia in Parkinson’s disease. II. Effects
168. Hordacre B, Goldsworthy MR, Vallence AM, et al. Variability in of subthreshold repetitive transcranial motor cortex stimulation.
neural excitability and plasticity induction in the human cortex: a Neurology 1994;44(5):892-898.
brain stimulation study. Brain Stim 2017;10(3):588-595. 189. Siebner HR, Mentschel C, Auer C, Conrad B. Repetitive trans-
169. Hannah R, Rocchi L, Tremblay S, Rothwell JC. Controllable pulse cranial magnetic stimulation has a beneficial effect on bradykinesia
parameter TMS and TMS-EEG as novel approaches to improve in Parkinson’s disease. Neuroreport 1999;10(3):589-594.
neural targeting with rTMS in human cerebral cortex. Front Neu- 190. Brusa L, Versace V, Koch G, et al. Low frequency rTMS of the
ral Circuits 2016;10:97. SMA transiently ameliorates peak-dose LID in Parkinson’s disease.
170. Maeda F, Keenan JP, Tormos JM, Topka H, Pascual-Leone A. Clin Neurophysiol 2006;117(9):1917-1921.
Interindividual variability of the modulatory effects of repetitive 191. Lomarev MP, Kanchana S, Bara-Jimenez W, Iyer M, Wassermann
transcranial magnetic stimulation on cortical excitability. Exp EM, Hallett M. Placebo-controlled study of rTMS for the treatment
Brain Res 2000;133(4):425-430. of Parkinson’s disease. Mov Disord 2006;21(3):325-331.
171. Hamada M, Murase N, Hasan A, Balaratnam M, Rothwell JC. 192. del Olmo MF, Bello O, Cudeiro J. Transcranial magnetic stimula-
The role of interneuron networks in driving human motor cortical tion over dorsolateral prefrontal cortex in Parkinson’s disease. Clin
plasticity. Cereb Cortex 2013;23(7):1593-1605. Neurophysiol 2007;118(1):131-139.

Movement Disorders, 2019 13


L A T O R R E E T A L

193. Benninger DH, Lomarev M, Wassermann EM, et al. Safety study 195. Gonzalez-Garcia N, Armony JL, Soto J, Trejo D, Alegria MA,
of 50 Hz repetitive transcranial magnetic stimulation in patients Drucker-Colin R. Effects of rTMS on Parkinson’s disease: a longi-
with Parkinson’s disease. Clin Neurophysiol 2009;120(4): tudinal fMRI study. J Neurol 2011;258(7):1268-1280.
809-815. 196. Mak MK. Repetitive transcranial magnetic stimulation combined
194. Kang SY, Wasaka T, Shamim EA, et al. Characteristics of the with treadmill training can modulate corticomotor inhibition and
sequence effect in Parkinson’s disease. Mov Disord 2010;25(13): improve walking performance in people with Parkinson’s disease.
2148-2155. J Physiother 2013;59(2):128.

14 Movement Disorders, 2019

View publication stats

You might also like