Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Jun 1.
Published in final edited form as: Parkinsonism Relat Disord. 2013 Mar 7;19(6):573–585. doi: 10.1016/j.parkreldis.2013.01.007

Safety of Transcranial Magnetic Stimulation in Parkinson’s Disease: A Review of the Literature

Matthew VonLoh 1, Robert Chen 2, Benzi Kluger 1,*
PMCID: PMC3653978  NIHMSID: NIHMS442027  PMID: 23473718

Abstract

Background

Transcranial magnetic stimulation (TMS) has been used in both physiological studies and, more recently, the therapy of Parkinson’s Disease (PD). Prior TMS studies in healthy subjects and other patient populations demonstrate a slight risk of seizures and other adverse events. Our goal was to estimate these risks and document other safety concerns specific to PD patients.

Methods

We performed an English-Language literature search through PudMed to review all TMS studies involving PD patients. We documented any seizures or other adverse events associated with these studies. Crude risks were calculated per subject and per session of TMS.

Results

We identified 84 single pulse (spTMS) and/or paired pulse (ppTMS) TMS studies involving 1091 patients and 77 repetitive TMS (rTMS) studies involving 1137 patients. Risk of adverse events was low in all protocols. spTMS and ppTMS risk per patient for any adverse event was 0.0018 (95% CI: 0.0002 – 0.0066) per patient and no seizures were encountered. Risk of an adverse event from rTMS was 0.040 (95% CI: 0.029 – 0.053) per patient and no seizures were reported. Other adverse events included transient headaches, scalp pain, tinnitus, nausea, increase in pre-existing pain, and muscle jerks. Transient worsening of Parkinsonian symptoms was noted in one study involving rTMS of the supplementary motor area (SMA).

Conclusion

We conclude that current TMS and rTMS protocols do not pose significant risks to PD patients. We would recommend that TMS users in this population follow the most recent safety guidelines but do not warrant additional precautions.

Keywords: Parkinson’s Disease, Transcranial Magnetic Stimulation, Safety

Introduction

Transcranial magnetic stimulation (TMS) is a noninvasive technique for cortical stimulation that uses electromagnetic induction to generate a strong fluctuating magnetic field which induces intracranial currents [1]. Single pulse (spTMS) and paired-pulse TMS (ppTMS) studies have been shown to be safe and effective in studying a variety of measures of motor cortex excitability including resting motor threshold, motor evoked potential amplitude, recruitment curves, cortical silent period, short interval intracortical inhibition, long interval intracortical inhibition and intracranial facilitation [2]. Studies of Parkinson’s Disease (PD) patients using these techniques have demonstrated that PD increases net cortical excitability and that effective therapeutic interventions including medications and surgery may reduce this excitability [3]. Repetitive TMS (rTMS) applies repeated TMS pulses at set frequencies or patterns to induce changes in cortical excitability which last longer than the period of stimulus administration [4]. These alterations have generally been observed as a decrease in cortical excitability with low-frequency stimulation (≤ 1 Hz) and an increase in cortical excitability with high frequency rTMS (≥ 5 Hz) [5]. Patterned rTMS protocols such as theta-burst stimulation (TBS) and repetitive paired-pulse stimulation utilize more complex trains of intermittent bursts and may induce even more durable alterations in cortical excitability [6].

rTMS has been investigated as a potential therapy for numerous conditions, including depression, epilepsy, migraine, and PD [79]. In PD, rTMS has been studied as an intervention to improve both motor symptoms, including rigidity and bradykinesia, motor complications of therapy (e.g. dyskinesias) and non-motor symptoms, including depression and speech [10]. In general, benefits when present have been of small to moderate magnitude and short-lived. However, given the potential for clinical benefit and limitations of medical options there is a need for further studies to further develop rTMS as a therapeutic intervention and to better define the longevity, efficacy, and benefit of rTMS [11].

The use of TMS in both healthy and clinical populations has been associated with several adverse events of varying severity. The most common are transient headaches and scalp discomfort. Scalp pain and headaches are thought to be due to activation of scalp pericranial muscles [2, 12]. However, more severe adverse effects may include mood changes (induction of mania), scalp burns from electrodes, and induction of seizures [2]. Seizures during TMS are thought to be a result of cortical pyramidal cell activation, spread of excitation to neighboring neurons, and overwhelming of inhibitory mechanisms [13]. Although reviews detailing the safety of TMS use exist for depression, epilepsy, and migraine, no such review exists for TMS use in PD [8, 14, 15]. Although PD is not associated with an increased risk of seizures, other neurophysiological changes may confer unique risks of TMS in the PD population including changes in cortical excitability and reductions in motor cortex inhibition.[16] Therefore, the purpose of this article is to provide a safety profile of TMS in PD for researchers and clinicians by reviewing the literature for any adverse events associated with TMS on PD patients.

Methods

Literature Review

A literature search for English language studies on TMS use in PD was conducted through PudMed. Review articles were excluded. The searches used included the following key words: transcranial magnetic stimulation, TMS, rTMS, Parkinson, Parkinson’s disease, silent period, Deep Brain Stimulation and theta burst. All applicable articles were reviewed for patient demographics (gender, age, medication status), TMS protocol used (TMS modality, method of localization, number of stimuli, stimuli intensity, coil type, and coil position) and adverse events reported. The review was conducted between 1992 and December 2011.

Statistical Analysis

We computed the proportion estimate of crude risk and 95% confidence intervals of seizures and other adverse events separately. We also separated single pulse and rTMS studies. Risks were calculated as per-person risk and per TMS session. Confidence intervals were calculated utilizing the Clopper-Pearson method in R software version 2.14.1. Fisher’s exact test was used to compare crude risks between groups.

Results

Single and Paired-Pulse TMS

We identified 84 studies utilizing single or paired pulse techniques in PD patients. This included 71 single-pulse protocols and 24 paired-pulse protocols including 1091 patients with PD [10, 1797]. Of these studies, 2 reported adverse events and 1 reported a transient change in motor performance. No seizures were reported, thus the crude risk of seizures is 0 (95% CI: 0.0000 – 0.0034). The risk of any adverse event during spTMS or ppTMS is 0.0018 (95% CI: 0.0002 – 0.0066) per patient.

Regarding adverse events potentially related to PD, Boylan et al. described a worsening of tremor in one patient following spTMS to the motor cortex during localization [98]. As this patient was also described to have an exaggerated startle response we suspect that the change in tremor may be more related to acute stress and not a specific physiologic reaction. Cunnington et al reported a transient increase in movement time required to complete a button pressing task in six patients following 100% maximum stimulator output (MO) spTMS of the SMA [62]. The slowing of movement only occurred when stimulation was administered early in the movement and was not found to be statistically correlated with patient age, severity of symptoms, or duration of disease. The authors hypothesized that this slowing reflected interruption of the SMA’s role in movement planning and is supported by other TMS research investigating the SMA in healthy populations.[99]

Regarding other adverse events, Benninger et al reported the occurrence of ipsilateral stimulation of cranial nerve (CN) VII in one patient following spTMS administered between trains of 50 Hz rTMS of M1, however the patient experienced no cranial nerve stimulation during the 50 Hz rTMS itself suggesting that this may be a coil placement issue [100].

rTMS

rTMS refers to repetitive TMS given either continuously at a low-frequency or in intermittent trains at higher frequencies. Theta Burst Stimulation (TBS) refers to a newer protocol where TMS stimulation is given in bursts of triplets at 50 Hz repeated in the theta range (5 Hz) either continuously (cTBS) or in ntermittent trains of 2 seconds (iTBS).[101] We identified 77 rTMS and TBS studies involving PD patients. This included 81 separate rTMS protocols and 8 TBS protocols involving a total of 1137 patients and 11672 rTMS sessions [10, 29, 30, 47, 51, 66, 80, 98, 100, 102164]. Tables 1 and 2 summarizes the demographic characteristics of these patients, study design, TMS parameters and any adverse events for rTMS and theta burst studies respectively. Of these studies, 14 reported the occurrence of an adverse event. There were no seizures reported. 51 adverse events were attributed to rTMS protocols. Of the 63 articles which did not report an adverse event, 33 protocols stated a lack of adverse events. The remaining 39 protocols neither stated nor denied the occurrence of any adverse events associated with rTMS or TBS. Out of 77 studies 4 reported scalp pain during treatment [98, 102, 118, 145], 5 reported mild transient headaches [106, 112, 117, 142, 145], plus 2 studies with an unstated number of headaches [106, 112, 117, 142, 145], 2 studies reported worsening performance of a motor task [98, 133], 1 TBS study reported transient (< 5 minutes) tinnitus [102], 1 study reported nausea [112], and 1 study reported transient increase in pre-existing back pain [113].

Table 1.

rTMS data

Author Year No. of
Subjects
On/Off
Medication
Age rTMS
Modailty
Method of
Localization
rTMS
frequency
No. of Stimuli
per Session
Intensity Coil Type Intertrain
interval
Session
schedule
Total Number
of Sessions
Target Adverse
events
Gonzalez-Garcia et al.[173] 2011 17 On 57 –70 high frequency NR 25 Hz 200 (M1); 2000 (occipital lobe) 80% RMT Fig8 NR 15 sessions over 3 months 255 M1; occipital lobe NR
Kodama et al.[154] 2011 1 On 45 low frequency Maximum MEP hotspot 0.9 Hz 200 (M1 hand); 300–600 (M1 leg) 110% AMT Fig8 NR 8 sessions over 2 months (M1 hand); 12 sessions over 3 month (M1 leg) 20 M1 hand; M1 leg None
Rektor et al.[163] 2010 10 NR NR low frequency NR 1 Hz 600 NR NR NR 1 session 10 DLPFC; IFC NR
Hartelius et al.[148] 2010 10 Off 39–67 high frequency Maximum MEP hotspot 10 Hz 2000 90% RMT Fig8 4 minutes 2 sessions over 2 consecutive days 10 M1 NR
Pal et al.[106] 2010 12 On/Off 59 –70 high frequency NR 5 Hz 600 90% RMT Fig8 20 s 10 sessions over 10 days 120 DLPFC Mild transient headache (n = 2)
Kang et al.[150] 2010 11 On/Off 48 –75 high frequency NR 25 Hz 1500 100% MT Fig8 10s 2 sessions 22 M1 NR
Arias et al. [138] 2010 a 9 On NR low frequency NR 1 Hz 100 90% RMT C 5 minute 10 sessions over 10 days 90 Vertex NR
Suppa et al.[160] 2010 14 On/Off 52 –77 high frequency Maximum MEP hotspot (M1); 2.5 cm anterior to the M1 hotspot (PMd) 5 Hz 1500 (PMd); 150 (M1) 90% RMT (PMd); 120% RMT (M1) Fig8 1 minute 2 sessions separated by 5 days 28 PMd; M1 None
9 On/Off 45 –63 high frequency Maximum MEP hotspot 5 Hz 450 120% RMT Fig8 1–2 minutes 1 session 9 M1 None
5 On 54 –73 high frequency Maximum MEP hotspot 1 Hz 1500 90% AMT Fig8 1 minute 2 sessions separated by at least 5 days 10 PMd None
Arias et al.[137] 2010b 9 On NR low frequency NR 1 Hz 100 90% RMT C 5 minute 10 sessions over 10 days 90 Vertex NR
Borgheres et al. [174] 2010 1 NR 79 high frequency NR 5 Hz 15 120% RMT Fig8 NR 1 session 1 M1 NR
Balaz et al.[139] 2010 18 On 55.8 +/−6.52 low frequency Frameless stereotaxy 1 Hz 600 80% RMT Fig8 NR 1 session 18 DLPFC (n = 8); IFC (n = 10) NR
Filipovic et al.[162] 2010 a 9 On 48 –73 low frequency Maximum MEP hotspot 1 Hz 1800 90% RMT Fig8 1 minute 4 sessions over 4 days 36 M1 None
Filipovic et al.[104] 2010 b 10 Off 49 –74 low frequency Maximum MEP hotspot 1 Hz 1800 90% RMT Fig8 1 minute 4 sessions over 4 days 40 M1 NR
Gruner et al.[105] 2010 15 On 56 –81 low frequency Maximum MEP hotspot 1 Hz 900 90% RMT Fig8 None 1 session 15 M1 None
Jacobs et al.[149] 2009 8 Off 62 +/−10 low frequency 5 cm anterior to the TA hotspot (SMA); 2.5 cm anterior to the FDI hotspot 1 Hz 1800 80% RMT Fig8 NR 2 sessions separated by 1 week 16 SMA; DLPFC NR
Furukawa et al.[147] 2009 6 On 62 –71 low frequency Maximum MEP hotspot 0.2 Hz 100 120% MT C NR 12 sessions over 3 months 72 M1 NR
Narayana et al.[108] 2009 1 On 59 high frequency Image-based robotically positioned TMS 4 Hz 400 110% MT NR 5s 10 sessions 10 M1 None
van Dijk et al.[161] 2009 13 On 46 –75 high frequency Maximum MEP hotspot (M1); 5 cm posterior to MEP hotspot (parietal cortex); 2.5 cm anterior to MEP hotspot (prefrontal cortex) 5 Hz 500 80% RMT Fig8 20s 10 sessions over 10 days 130 parietal cortex (n = 8); M1 or premotor cortex (n = 7) None
Baumer et al.[109] 2009 15 On/Off 63.1 +/−6.8 low frequency Maximum MEP hotspot 1 Hz 1200 80% AMT Fig8 NA 4 sessions 60 PMd NR
Benninger et al.[100] 2009 10 On 50 –77 high frequency Maximum MEP hotspot 50 Hz 1000 60% –90% RMT C NR 1 session 10 M1 None
Sedlackoa et al.[110] 2009 10 Off 52 –79 high frequency Frameless stereotaxy 10 Hz 1350 100% RMT Fig8 10 s 3 sessions separated by 10 min 30 DLPFC; occipital cortex; dorsal premotor cortex None
Rothkegel et al.[112] 2009 22 On/Off 34 –76 low frequency Maximum MEP hotspot 0.5 Hz 600 80% RMT Fig8 NA 1 session 22 M1 Headache (n=2), nausea (n=1)
high frequency Maximum MEP hotspot 10 Hz 2000 80% RMT Fig8 50s 1 session 22 M1
Brusa et al.[143] 2009 8 On 52 –75 low frequency 1 cm anterior to Cz 1 Hz 900 65% MO Fig8 NA 10 sessions over 2 weeks 80 M1 NR
Cardoso et al.[142] 2008 11 Off 67 +/−8.3 high frequency 5 cm anterior to optimal stimulation of abductor pollicis brevis 5 Hz 3,750 120 % MT Fig8 NR 12 sessions over 4 weeks 132 DLPFC Headache (equally distributed in both rTMS and rTMS sham groups
Filipovic et al.[175] 2008 5 On 48 –74 low frequency Maximum MEP hotspot 1 Hz 1800 90% AMT Fig8 1 minute 4 sessions over 4 days 20 M1 None
Rodrigues et al. [47] 2008 6 On/Off 62 –73 low frequency Maximum MEP hotspot 0.2Hz 440 130% RMT Fig8 5s 2 sessions, 1 on and 1 off medication 12 M1 None
Hamada et al.[113] 2008 55 On 39 –82 high frequency 3-cm anterior to maximum MEP hotspot for tibialis anterior 5 Hz 1000 110% AMT Fig8 50s 8 sessions over 8 weeks 440 SMA Lower back pain increased (n = 1)
Rektorova et al.[114] 2008 6 On 67.3 +/−7.7 high frequency Optimum activation of FDI or TA 10 Hz 1350 90% RMT C NR 5 sessions over 5 consecutive days 30 DLPFC None
Fierro et al.[66] 2008 14 On/Off 48 –82 high frequency Maximum MEP hotspot 10 Hz 500 90% MT Fig8 30s 2 sessions 28 M1 NR
Kim et al.[152] 2008 9 Off 43 –68 high frequency NR 5 Hz 75 90% RMT Fig8 10s 2 sessions over 2 consecutive days 18 M1 NR
Epstein et al.[115] 2007 14 On/Off 42 –78 high frequency MEP w/lowest threshold 10 Hz 1000 110% RMT Custom iron core coil 25s 20 sessions over 10 days 280 M1 None
Kormos [176] 2007 7 Off 62 –79 high frequency NR 20 Hz 2000 80% MT NR 28s 10 sessions over 2 weeks 70 DLPFC None
Rektorova et al.[157] 2007 6 On 63.7 +/−7.7 high frequency NR 10 Hz 1350 90% RMT Fig8 NR 5 sessions over 5 days 30 MC, DLPFC NR
Khedr et al.[151] 2007 22 Off 45 –85 high frequency NR 25 Hz 2000 100% RMT Fig8 50s 36 sessions over 6 days 792 M1 NR
Anninos et al.[116] 2007 30 Off 49 –80 high frequency NR 8 –13 Hz 2880 –4680 1–7.5 pT C NR 3 sessions, 1 in lab and 2 self-administere d at patient’s home 90 Left and right temporal regions, frontal and occipital regions, vertex None
Loscher et al.[155] 2007 8 On 58.5 +/−5.3 high frequency NR 5 Hz 100 MEP = 0.5 –1mV Fig8 1 minute 1 session 8 M1 NR
Del Olmo et al.[144] 2007 8 On 54 –74 high frequency 5 cm anterior to maximum MEP for FDI 10 Hz 450 90% RMT Fig8 10s 10 sessions over 10 days 80 DLPFC NR
Fregni et al. [136] 2006 13 On 65.2 +/−7.9 high frequency 5 cm anterior to maximum MEP for APB 15 Hz 3000 110% RMT Fig8 10s 10 sessions over 2 weeks 130 DLPFC NR
Brusa et al.[141] 2006 10 Off 61 +/−8.04 low frequency 3 cm anterior to Cz 1 Hz 900 90% RMT Fig8 NA 2 sessions 20 SMA None
2006 10 On 61 +/−8.04 low frequency 3 cm aterior to Cz 1 Hz 900 90% RMT Fig8 NA 5 sessions over 5 days 50 SMA None
Cincotta et al.[29] 2006 3 NR 60 –82 high frequency Maximum MEP hotspot 5Hz 15 120% RMT Fig8 NA 4 sessions 12 M1 NR
Morgante et al.[30] 2006 16 On/Off 50 –80 low frequency Maximum MEP hotspot 0.1 Hz 20 MEP = 1mV Fig8 NA 6 sessions, 3 on medication and 3 off medication 96 M1 NR
Khedr et al.[117] 2006 55 Off 30 –85 high frequency NR 10/25 Hz 2000 100% MT Fig8 50 s 36 sessions, 6 sessions per day for 6 days 1980 Bilateral M1 for lower limbs, Bilateral M1 for the hand Mild, transient headache in some patients
Lomarev et al.[118] 2006 18 On 63 +/−10 high frequency NR 25 Hz 1200 100% MT Fig8 NR 8 sessions over a 4-week period 144 Left and right motor and DLPFC Intolerable pain (n=1)
Dias et al.[64] 2006 11 On 68.47 +/−4.75 high frequency Maximum MEP hotspot 15 Hz 3000 110% MT Fig8 10s 10 sessions over 2 weeks 110 DLPFC None
2006 8 On 61.31 +/−8.46 high frequency Maximum MEP hotspot 5 Hz 2250 90% MT Fig8 5s 1 session 8 M1 None
Strafella et al.[119] 2005 7 Off 40 –66 high frequency MEP w/lowest threshold 10 Hz 600 90% RMT C 10s 2 sessions over 2 days 14 M1 NR
Boggio et al. [120] 2005 13 Off NR high frequency Maximum MEP hotspot 15 Hz 3000 110% MT Fig8 NR 10 sessions over 2 weeks 130 Left DLPFC None
Koch et al.[153] 2005 8 Off 48 –73 low frequency 3 cm anterior to Cz 1 Hz 900 90% RMT Fig8 NA 1 session 8 SMA NR
high frequency 3 cm anterior to Cz 5 Hz 900 110% RMT Fig8 40s 1 session 8 SMA NR
Mir et al.[80] 2005 9 On/Off 47 –73 high frequency Maximum MEP hotspot 5 Hz 1500 90% AMT Fig8 1 minute 2 sessions 18 PMd None
Buhmann et al.[51] 2004 16 On/Off 58.4 +/−10.5 low frequency Maximum MEP hotspot 1 Hz 1200 80% AMT Fig8 NA 2 sessions over 2 weeks 32 PMd None
Lefaucheur et al.[10] 2004 12 Off 51 –76 low frequency Maximum MEP hotspot 0.5 Hz 600 80% RMT Fig8 NA 1 session 12 M1 None
high frequency Maximum MEP hotspot 10Hz 2000 80% RMT Fig8 50s 1 session 12 M1 None
Mally et al.[156] 2004 46 On 63.9 +/−9 low frequency NR 1 Hz 50 25% MO (MO = 2.3T) C NA 42 sessions over 3 years administered in 7 sessions over 7 days 1932 Vertex None
Fregni et al. [121] 2004 21 On 50 –80 high frequency NR 15 Hz 3000 110% MT Fig8 NR 10 sessions over 10 days 210 Left DLPFC NR
Koch et al.[122] 2004 20 Off 61 +/−6.83 high frequency 3 cm anterior to vertex (SMA), Intersection of coil loops at F4 (DLPFC) 5 Hz 250 100% MT Fig8 30s 2 sessions on 2 separate days 40 SMA and right DLPFC NR
Bornke et al.[140] 2004 12 Off 37 –74 high frequency NR 10 Hz 1000 90% RMT Fig8 10s 2 sessions over 4 days 24 M1 None
Ikeguchi et al.[123] 2003 12 On 51 –78 low frequency F3 or F4 of the international 10–20 system 0.2 Hz 30 70% MO C NA 6 sessions over 2 weeks 72 Frontal (L middle frontal gyrus, R inferior frontal gyrus); Occipital (L lingual gyrus, R posterior lobe of cerebellum) None
Khedr et al.[124] 2003 19 Off 36 –70 high frequency Maximum MEP hotspot 5 Hz 2000 120% MT Fig8 NR 10 sessions over 10 days 190 M1 (EDB) 1000 pulses; M1 (hand) 500 pulses/hemisphere NR
Okabe et al.[125] 2003 85 (1/3 received sham) On 67.2 +/−8.2 low frequency NR 0.2 Hz 100 110% AMT C NA 8 sessions over 8 weeks 680 M1 and occipital cortex NR
Gilio et al. [126] 2002 15 On/Off (4 patients only off; the rest off/on) 46 –76 high frequency NR 5 Hz 40 (Off/On medication ); 160 (Off medication only) 120% RMT Fig8 1 minute 2 sessions in 1 day 30 M1 NR
Sommer et al.[127] 2002 11 On 35 –77 low frequency Maximum MEP hotspot 1 Hz 900 120% RMT Fig8 NA 3 sessions over 3 days 33 M1 None
Dragasevic et al.[145] 2002 10 On 46 –72 low frequency 6 cm anterior to point of motor threshold determination 0.5 Hz 100 110% MT C 1 minute 20 sessions over 10 days 200 Prefrontal area Light burning sensations over the scalp (n = 4); mild tension headache (n = 3)
Boylan et al.[98] 2001 10 Off 55 –77 high frequency Visible muscle twitch 10 Hz 2000 110% MT, 68–78% MT for 3 patients Fig8 55s 1 session 10 SMA Scalp discomfort at 110% maximum MEP (n = 3); Subclinical worsening of complex and preparatory movement (spiral drawing) following rTMS to SMA (n = 5)
Shimamoto [128] 2001 9 On 53 –79 low frequency NR 0.2 Hz 60 78% MO (700V) C NA 8 sessions over 8 weeks 72 Frontal area NR
Siebner et al.[129] 2000 a 10 Off 57 +/−11 high frequency Maximum MEP hotspot 5 Hz 2250 90% RMT Fig8 10s 1 session 10 M1 None
Siebner et al.[130] 2000b 10 Off 41–75 high frequency Maximum MEP hotspot 5 Hz 2250 130% RMT Fig8 10s 1 session 10 M1 NR
Tergau et al[164] 1999 7 On 54 –73 low frequency Maximum MEP hotspot 1 Hz 500 90% MT C NA 1 session 7 M1 NR
high frequency Maximum MEP hotspot 5 Hz 500 90% MT C 30s 1 session 7 M1 NR
high frequency Maximum MEP hotspot 10 Hz 500 90% MT C 20s 1 session 7 M1 NR
high frequency Maximum MEP hotspot 20 Hz 500 90% MT C 45s 1 session 7 M1 NR
Mally et al.[131] 1999 a 49 On NR low frequency NR 1Hz 30, 60 15 –30% MO C NA 10 sessions over 10 days, 14 sessions over 14 days 1176 Vertex None
Ghabra et al.[133] 1999 11 Off 48 –70 high frequency Maximum MEP hotspot 5 Hz NR 90% RMT Fig8 NR 2 sessions 22 M1 Muscle jerks during motor task (n=11)
Mally et al. [132] 1999b 10 On 56 –73 low frequency NR 1 Hz 30 20% MT C NR 20 sessions over 10 days 200 Vertex NR
Siebner et al.[134] 1999 12 Off 41 –74 high frequency Maximum MEP hotspot 5 Hz 2250 90% RMT Fig8 10 s 2 sessions over 2 days 24 M1 None
Sandyk [158] 1998 2 On 49, 73 high frequency NR 5, 7 Hz 6000, 8400 7.5 pT NR NR 4 5 Hz and 4 7 Hz sessions over 4 days 16 NR NR
Pascual-Leone et al.[177] 1994 6 On/Off 48 –73 high frequency Maximum MEP hotspot 5 Hz NR 10% RMT Fig8 NR 3 sessions 18 M1 None
Totals 1068 11,198 17 scalp pain, 12 mild transient headaches, 1 study with an unstated number of headaches, 16 worsening performance of a motor task, 1 nausea, and 1 transient increase in pre-existing back pain

Table 2.

Theta Burst Stimulation Studies

Author Year Number of Subjects On/Off Medication Age TMS Parameters Adverse Events
Stephani et al.[166] 2011 8 On 62.2 ± 8.3 3 sessions at least one week apart of M1 iTBS, sham iTBS and tRNS given at 80% rMT for 10 minutes. NR
Benninger et al.[102] 2011 13 On 62.1 ± 6.9 8 sessions over two consecutive weeks of iTBS over bilateral M1 and DLPFC at 80% aMT for 600 pulses per site per session and 4800 total pulses. Transient tinnitus (< 5 minutes, N = 1) and occasional local pain during stimulation
Suppa et al.[178] 2011 20 On 48–76 1 session of iTBS over M1 at 80% aMT for a total of 600 pulses. No adverse effects.
Eggers et al.[167] 2010 8 Off 60–78 One session of cTBS over m1 at 80% aMT for a total of 600 pulses. NR
Koch et al.[111] 2009 20 On 64.2 ± 5.4 10 sessions of bilateral cerebellar cTBS at 80% aMT f0r 600 pulses per side per session and 12,000 total pulses. No Adverse Effects.
Rothkegel et al.[168] 2008 22 Both 34–76 5 sessions on 5 consecutive days over M1 including sham iTBS (600 pulses), high frequency rTMS (10 Hz for 2000 pulses at 80%rMT), low frequency rTMS (0.5 Hz at 80% rMT for 600 pulses), cTBS (600 pulses at 80% aMT) and iTBS (600 pulses at 80% aMT) NR
 Total 91 1 episode transient tinnitus; unspecified number with occasional local pain

aMT – active motor threshold; cTBS –continuous theta burst stimulation; DLPFC – dorsolateral prefrontal cortex; iTBS – intermittent theta burst stimulation; M1 – motor cortex; rMT – resting motor threshold; tRNS – Transcranial random noise stimulation

The crude risk of seizures in PD subjects is thus 0 (95% CI: 0 – 0.0032) per person and 0 (95% CI: 0 – 0.0003 ) per rTMS or TBS session. The crude risk of other adverse events in PD subjects is 0.040 (95% CI: 0.029 – 0.053) per person and 0.0039 (95% CI: 0.0028 – 0.0052) per rTMS or TBS session. Comparing protocols with a single session (N = 380) to those with multiple sessions (N = 688) reveals a significant increase in risk with multiple sessions (Fisher’s exact test, p < 0.001) suggesting that risk is at least partially cumulative over sessions rather than an all or none occurrence for certain high-risk subjects.

Regarding adverse events potentially related to PD, motor symptoms were shown to worsen of selected motor tasks in patients following certain rTMS protocols (N = 16). Boylan et al. reported worsening of spiral drawing in five patients following 10 Hz rTMS of the SMA [98]. This finding may relate to the role of the SMA in movement preparation as demonstrated in control subjects. Ghabra et al. reported muscle jerks during 90%, RMT 5 hz rTMS over M1 such that eleven patients could not complete a concurrent Grooved Pegboard task. This “jerking” likely reflected MEPs induced with a lowering of motor threshold when subjects activated motor cortex during the skilled motor task. Upon rTMS intensity reduction to 75–85% RMT all patients were able to complete the task. One patient in this study also noted a worsening of action tremor at the higher stimulation intensity which resolved at 75% RMT rTMS intensity and may reveal a potential interaction between motor cortex activation, whether external or internal, and action tremor.

Regarding adverse events not related to PD, the most common adverse effects reported were headache (N = 7) and local pain (N = 17). Authors gave the following descriptions of adverse events. Pal et al. reported the occurrence of mild transient headache in two patients which required neither interruption of study or medication attention following 5 Hz rTMS of M1 [106]. Rothkegel et al. reported headache in two patients following TMS of M1, though the modality which caused the side effects was not specified out of the four used (rTMS at 0.5 Hz and 10 Hz, iTBS, and cTBS) [112]. Cardoso et al. reported an unspecified number of headaches which were spread equally amongst the rTMS group and the sham rTMS group using a sham coil [142]. Khedr et al. reported the occurrence of mild transient headache following 25 Hz rTMS of M1, though an exact number of patients experiencing the event was not stated [117]. Dragasevic et al. reported mild tension headache in 3 patients following 0.5 Hz rTMS of the prefrontal area [145]. Boylan et al. reported scalp discomfort (N = 3) following 10 Hz rTMS of SMA [98]. Benninger et al. reported scalp pain associated with DLPFC stimulation in nine subjects following intermittent theta-burst stimulation (iTBS) of the primary motor cortex (M1) [102]. [100]. Lomarev et al. reported intolerable pain located under the coil position in one patient following 25 Hz rTMS of M1 and dorsolateral prefrontal cortex, due to which the patient dropped out of the study [118]. Dragasevic et al. reported light burning sensations over the scalp in four patients following 0.5 Hz rTMS of the prefrontal area [145]. Boylan et al. reported scalp discomfort in three patients following 10 Hz rTMS of the SMA which was alleviated by reducing the stimulus intensity from 110% motor threshold (MT) to 68% – 78% MT [98].

Other adverse events reported included tinnitus (N = 1), nausea (N = 1), and an increase in previously acquired lower back pain (N = 3). Benninger et al. reported a nonpulsatile left-sided tinnitus for a few minutes in one subject following intermittent theta-burst stimulation (iTBS) of the primary motor cortex (M1) [102]. Rothkegel et al. reported nausea in one patient following TMS of M1, though the modality which caused the side effects was not specified out of the four used (rTMS at 0.5 Hz and 10 Hz, iTBS, and cTBS) [112]. Hamada et al. reported an increased sensation of back pain which existed prior to treatment in one patient following 5 Hz rTMS of the supplementary motor area (SMA) [113].

A number of events which either did not directly result in negative outcomes for the patient or were not attributed to the rTMS procedure were also reported. Due to this, these events were not included in the risk assessments, but are included here for completeness. Beninnger et al. reported one patient with residual muscle activity and possible spread of excitation from arm to lower extremity muscles by clinical observation following 50 Hz rTMS.[100] This subject also had a slight increase in left temporal spikes monitored by electroencephalography (EEG) but had occasional bitemporal spikes at baseline and upon further questioning after the rTMS session mentioned a prior car accident with blunt head trauma and possible loss of consciousness. Epstein et al. reported the occurrence of falls (n=4), a recurrence of paroxysmal atrial fibrillation (n=1), and unilateral hip pain unrelated to any acute injury (n=1) during a trial of 10 Hz rTMS of M1. However these events were not temporally related to the rTMS and thus not considered side effects of rTMS treatment [115]. Mally et al. reported the occurrence of dystonia in four patients which was thought to be a result of drug treatment with levodopa and extended release levodopa and not a result of 1 Hz rTMS at the vertex [131].

Sham TMS was used in both rTMS[104, 106, 113, 125, 128, 142, 144, 150, 162, 165] (N = 142) and TBS[102, 111, 166168] (N = 58) protocols. Of these sham exposures, one patient receiving sham rTMS over SMA withdrew due to perceived worsening of symptoms[113] and one study reported a similar incidence of mild headaches in their real and sham 5 Hz DLPFC rTMS groups.[142] While the number of adverse events for both real and sham rTMS are small, Fisher’s exact test (P > 0.05) does not reveal a significant difference between crude rates of side effects and suggests that caution may be warranted when attributing side effects observed in studies to the physiological effects of rTMS.

TMS in Patients with Deep Brain Stimulators

In 1999 Kumar et al. tested TMS pulses delivered over DBS leads embedded in conduction gel and directly over stimulators to demonstrate that TMS in DBS patients did not effect DBS leads but could disrupt stimulator function if stimulated directly over the stimulator device.[169] Since that time there have been a number of studies using TMS in PD patients following deep brain stimulation (DBS) surgery of the subthalamic nucleus (STN; see Table 3) with no adverse events reported in 122 subjects. The crude risk of any adverse event in PD STN DBS subjects is thus 0 (95% CI: 0 – 0.0298) per person. While only one of these studies included patients who also had globus pallidus interna (GPI) DBS,[170] studies in dystonia subjects with GPI DBS would suggest that these patients would also be reasonable candidates for future DBS research.[171]

Table 3.

TMS Use in PD Patient’s with STN DBS Devices

Author Year Number of Subjects On/Off Medication On/Off DBS Age TMS Parameters Adverse Events
Balaz et al.[165] 2010 18 On Off 55.8 ± 6.5 1 session of rTMS at 80% rMT at 1 Hz over either IFC or DLPFC for 600 total pulses NR
Kuriakose et al.[179] 2010 8 On Both 52–75 1 session of single pulse TMS over M1 delivered every 6 seconds in 3 different coil orientations (AP, PA and perpendicular) following DBS pulses for approximately 180 total pulses. NR
Rektor et al.[180]* 2010 10 NR Off NR 1 session of 1 Hz rTMS for 600 pulses over either right IFC or DLPFC; Intensity NR NR
Baumer et al.[181] 2009 15 Both Both 60.3 ± 6.3 4 single pulse TMS sessions over M1 consisting of rMT determination and 10 pulses at 150% of rMT over motor hotspot for SP determination NR
Narayana[182] 2009 1 NR Off 59 10 sessions of 4Hz rTMS over left PMd at 110% rMT delivered in 5 second trains with a 5 second intertrain interval. 20 trains were given per session for a total of 4000 pulses. No Adverse Effects. TMS mimicked aspects of DBS induced speech dysfunction which was the intended effect (virtual lesion).
Gaynor et al.[183] 2008 9 On Off 50–69 1 session including 30–50 single pulses every 5 seconds over one or both M1 and left SMA at 95% and 115% rMT NR
Fraix et al.[184] 2008 15 Off Both 60 ± 11 1 session over M1 of single pulse (SP, rMT, aMT, CMCT) and paired –pulse (SICI, ICF) measures for approximately 180 total pulses. No adverse effects.
Potter-Nerger et al.[185] 2008 10 Off Both 58.3 ± 8.3 1 session of 95% aMT single pulses over M1 (soleus hotspot) for approximately 60 total pulses. NR
Sailer et al.[186] 2007 7 Both Both 56.1 ± 6.3 1 session of single pulse TMS at rMT over M1 paired with median nerve stimulation to measure SAI and LAI for approximately 80 total pulses. NR
Compta et al.[187] 2006 3 Off Off NR 1 session of suprathreshold SP determination for approximately 10 total pulses. NR
Hidding et al.[188] 2006 8 Off Off 43–69 1 session of RC and CMCT over M1 at 110%–150% rMT for approximately 50 total pulses. NR
Kuhn et al.[189] 2004 5 On Off 56.8 ± 3.0 1 session of single pulse TMS over M1 above rMT with or without acoustic stimulation. Estimated 20–50 total pulses. NR
Dauper et al.[53] 2002 8 Both Both 59.3 ± 10.0 4 sessions of single pulse (MEP, SP) and paired-pulse (SICI, ICF) at 120% rMT for approximately 80 total pulses. No adverse events.
Cunic et al.[40] 2002 9 On Both 41–78 3 sessions of single pulse (MT, RC, SP) and paired-pulse (SICI, LICI, ICF) M1 stimulation at 100–150% rMT at rest and active contraction for estimated 360 total pulses. No adverse effects.
Pierantozzi et al.[170] 2002 4 (implanted in both bilateral STN and GPI) Both Both 49–60 4 sessions of single pulse (rMT) and paired-pulse (SICI) at 70%–120% rMT for approximately 120 total pulses. NR
Total 122 NR

Abbreviations: aMT – active motor threshold; CMCT – central motor conduction time; DBS - Deep Brain Stimulation; ICF – intracortical facilitation; LAI – long latency afferent inhibition; M1 – primary motor cortex; MEP – motor evoked potential; NR – not reported; PMd – dorsal premotor cortex; RC – recruitment curve; rMT – resting motor threshold; rTMS – repetitive transcranial magnetic stimulation; SAI – short latency afferent inhibition; SICI – short intracortical inhibition; SMA – supplemental motor area; SP – silent period; STN - Subthalamic Nucleus; TMS – Transcranial Magnetic Stimulation

*

May overlap patients in Balaz study

Conclusions

TMS has been shown to be a useful technique for studying the neurophysiology of PD and shows potential in the treatment of motor and non-motor symptoms. Our review of the literature, including 2228 patients, revealed that both TMS and rTMS do not carry significant risk of adverse events in the PD population. Based on our review, we would suggest that TMS and rTMS may have similar risks to those found in the general population and that these risks, while low, do increase over multiple sessions. We would recommend that TMS users in this population follow the most recent safety guidelines but do not warrant additional precautions. We would however recommend that rTMS studies in PD patients monitor for motor function, particularly with SMA stimulation. We would also recommend that EEG and EMG monitoring be utilized for novel stimulation paradigms, as exemplified by Benninger et al. but do not feel that this level of monitoring needs to be used routinely.[100] Finally, preliminary evidence from 122 PD patients with DBS implants similarly suggests that TMS does not carry a significant risk in this population either.

One unique issue raised in this review is the potential for worsening motor symptoms with certain spTMS and rTMS paradigms [62, 98]. The Cunnington et al. spTMS study’s findings of increased time to complete a movement was attributed to a disturbance of the SMA’s role in motor planning due to the occurrence of the adverse event only when administered early in the movement [62]. Detrimental effects on spiral drawing and the preparatory phase of movement due to physiological disturbance of SMA has been observed in studies prior to Boylan et al., including the Cunnington et al. study on PD patients [62, 172]. The Boylan et al. study suggests that rTMS may be able to make such disruptions persist beyond the initial stimulus [98]. However, Hamada et al. found that SMA stimulation resulted in improvement of motor symptoms in PD patients as measured by UPDRS scores [113]. There are several possible causes for the difference between the two study’s findings. Hamada et al used a 5 Hz stimulation frequency as compared to Boylan et al. using 10 Hz [98, 113]. In addition, Hamada et al. delivered only 1000 stimuli per session, while Boylan et al. delivered 2000 stimuli [98, 113]. The increase in rTMS intensity and total number of stimuli may have caused Boylan et al. to elicit a negative outcome due to excessive excitation of the SMA. Another potential difference lies in the time course of the two studies. Boylan et al. only delivered 2 sessions at least one week apart [98]. Hamada et al. however did not see improvement of motor symptoms in their patients until at least 4 consecutive weeks of rTMS treatment [113]. Thus it is possible that reduction of risk and presence of benefit in rTMS of the SMA will only be achieved by lower intensity treatment over a longer timeframe. The conflicting results between these two studies merit further investigation of rTMS stimulation of the SMA in PD patients. We therefore recommend that rTMS studies in PD patients monitor for motor fluctuations and worsening.

All other adverse events attributed to rTMS were minor and no studies reported the need for medical care in response an event. Out of 1137 patients 17 reported scalp pain during treatment [98, 102, 118, 145], 7 reported mild transient headaches, plus 2 studies with an unstated number of headaches [106, 112, 117, 142, 145], 1 reported transient tinnitus [102], 1 reported nausea [112], and 1 reported transient increase in pre-existing back pain [113]. Due to their low rate of occurrence, transient nature, and complete lack of need for medical intervention these adverse events can be considered of minimal risk to the patient.

A further caveat concerns other potential risks in the PD population. First, medications should be carefully screened to ensure that medications associated with a lowered seizure threshold (e.g. antipsychotics, psychostimulants, tricyclic antidepressants, buproprion) are either excluded or carefully monitored. This would include antipsychotics and certain antidepressants. Second, PD patients should be screened as other patients for associated comorbidities including cardiac disease and epilepsy. Finally, patients with vascular Parkinsonism may have an increased risk of seizure.

We conclude that established TMS protocols have a minimal risk of adverse events in the PD patient population. PD patients should still be warned of the potential risk for seizure due to rTMS in the general population as well as a small risk of transient headache and scalp pain seen in previous PD study participants. However, the use of TMS should be encouraged in the further study of the neuronal processes underlying PD as well as an alternative treatment for PD so long as it is thought to produce clinically relevant improvements in motor function.

Acknowledgments

This work was supported by the National Institutes of Health/Colorado Clinical Translational Sciences Institute (5 KL2 RR025779-03; BMK). The authors would like to thank Alice Liu and Collette Celani-Morrell for her assistance in the preparation of this manuscript

Funding agencies: National Institutes of Health

Footnotes

Relevant conflicts of interest/financial disclosures: Nothing to report.

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

References

  • 1.Kobayashi M, Pascual-Leone A. Transcranial magnetic stimulation in neurology. Lancet Neurol. 2003;2(3):145–56. doi: 10.1016/s1474-4422(03)00321-1. [DOI] [PubMed] [Google Scholar]
  • 2.Wassermann EM. Risk and safety of repetitive transcranial magnetic stimulation: report and suggested guidelines from the International Workshop on the Safety of Repetitive Transcranial Magnetic Stimulation, June 5–7, 1996. Electroencephalogr Clin Neurophysiol. 1998;108(1):1–16. doi: 10.1016/s0168-5597(97)00096-8. [DOI] [PubMed] [Google Scholar]
  • 3.Berardelli A. Transcranial magnetic stimulation in movement disorders. Electroencephalogr Clin Neurophysiol Suppl. 1999;51:276–80. [PubMed] [Google Scholar]
  • 4.Anand S, Hotson J. Transcranial magnetic stimulation: neurophysiological applications and safety. Brain Cogn. 2002;50(3):366–86. doi: 10.1016/s0278-2626(02)00512-2. [DOI] [PubMed] [Google Scholar]
  • 5.Pascual-Leone A, et al. The plastic human brain cortex. Annu Rev Neurosci. 2005;28:377–401. doi: 10.1146/annurev.neuro.27.070203.144216. [DOI] [PubMed] [Google Scholar]
  • 6.Pell GS, Roth Y, Zangen A. Modulation of cortical excitability induced by repetitive transcranial magnetic stimulation: influence of timing and geometrical parameters and underlying mechanisms. Prog Neurobiol. 2011;93(1):59–98. doi: 10.1016/j.pneurobio.2010.10.003. [DOI] [PubMed] [Google Scholar]
  • 7.Howland RH, et al. The emerging use of technology for the treatment of depression and other neuropsychiatric disorders. Ann Clin Psychiatry. 2011;23(1):48–62. [PubMed] [Google Scholar]
  • 8.Bae EH, et al. Safety and tolerability of repetitive transcranial magnetic stimulation in patients with epilepsy: a review of the literature. Epilepsy Behav. 2007;10(4):521–8. doi: 10.1016/j.yebeh.2007.03.004. [DOI] [PubMed] [Google Scholar]
  • 9.Lipton RB, Pearlman SH. Transcranial magnetic simulation in the treatment of migraine. Neurotherapeutics. 2010;7(2):204–12. doi: 10.1016/j.nurt.2010.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Lefaucheur JP, et al. Improvement of motor performance and modulation of cortical excitability by repetitive transcranial magnetic stimulation of the motor cortex in Parkinson’s disease. Clin Neurophysiol. 2004;115(11):2530–41. doi: 10.1016/j.clinph.2004.05.025. [DOI] [PubMed] [Google Scholar]
  • 11.Fregni F, et al. Non-invasive brain stimulation for Parkinson’s disease: a systematic review and meta-analysis of the literature. J Neurol Neurosurg Psychiatry. 2005;76(12):1614–23. doi: 10.1136/jnnp.2005.069849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Wassermann EM, et al. Repetitive transcranial magnetic stimulation of the dominant hemisphere can disrupt visual naming in temporal lobe epilepsy patients. Neuropsychologia. 1999;37(5):537–44. doi: 10.1016/s0028-3932(98)00102-x. [DOI] [PubMed] [Google Scholar]
  • 13.Daskalakis ZJ, et al. Reduced cerebellar inhibition in schizophrenia: a preliminary study. Am J Psychiatry. 2005;162(6):1203–5. doi: 10.1176/appi.ajp.162.6.1203. [DOI] [PubMed] [Google Scholar]
  • 14.Machii K, et al. Safety of rTMS to non-motor cortical areas in healthy participants and patients. Clin Neurophysiol. 2006;117(2):455–71. doi: 10.1016/j.clinph.2005.10.014. [DOI] [PubMed] [Google Scholar]
  • 15.Dodick DW, et al. Transcranial magnetic stimulation for migraine: a safety review. Headache. 2010;50(7):1153–63. doi: 10.1111/j.1526-4610.2010.01697.x. [DOI] [PubMed] [Google Scholar]
  • 16.Lefaucheur JP. Motor cortex dysfunction revealed by cortical excitability studies in Parkinson’s disease: influence of antiparkinsonian treatment and cortical stimulation. Clinical neurophysiology: official journal of the International Federation of Clinical Neurophysiology. 2005;116(2):244–53. doi: 10.1016/j.clinph.2004.11.017. [DOI] [PubMed] [Google Scholar]
  • 17.Vacherot F, et al. A motor cortex excitability and gait analysis on Parkinsonian patients. Mov Disord. 2010;25(16):2747–55. doi: 10.1002/mds.23378. [DOI] [PubMed] [Google Scholar]
  • 18.Ni Z, et al. Involvement of the cerebellothalamocortical pathway in Parkinson disease. Ann Neurol. 2010;68(6):816–24. doi: 10.1002/ana.22221. [DOI] [PubMed] [Google Scholar]
  • 19.Kuriakose R, et al. The nature and time course of cortical activation following subthalamic stimulation in Parkinson’s disease. Cereb Cortex. 2010;20(8):1926–36. doi: 10.1093/cercor/bhp269. [DOI] [PubMed] [Google Scholar]
  • 20.Potter-Nerger M, et al. Subthalamic nucleus stimulation restores corticospinal facilitation in Parkinson’s disease. Mov Disord. 2008;23(15):2210–5. doi: 10.1002/mds.22284. [DOI] [PubMed] [Google Scholar]
  • 21.Schneider SA, et al. Motor cortical physiology in patients and asymptomatic carriers of parkin gene mutations. Mov Disord. 2008;23(13):1812–9. doi: 10.1002/mds.22025. [DOI] [PubMed] [Google Scholar]
  • 22.Gaynor LM, et al. Suppression of beta oscillations in the subthalamic nucleus following cortical stimulation in humans. Eur J Neurosci. 2008;28(8):1686–95. doi: 10.1111/j.1460-9568.2008.06363.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Schrader C, et al. Changes in processing of proprioceptive information in Parkinson’s disease and multiple system atrophy. Clin Neurophysiol. 2008;119(5):1139–46. doi: 10.1016/j.clinph.2008.01.005. [DOI] [PubMed] [Google Scholar]
  • 24.Shin HW, Kang SY, Sohn YH. Disturbed surround inhibition in preclinical parkinsonism. Clin Neurophysiol. 2007;118(10):2176–9. doi: 10.1016/j.clinph.2007.06.058. [DOI] [PubMed] [Google Scholar]
  • 25.Wu AD, et al. Asymmetric corticomotor excitability correlations in early Parkinson’s disease. Mov Disord. 2007;22(11):1587–93. doi: 10.1002/mds.21565. [DOI] [PubMed] [Google Scholar]
  • 26.Eusebio A, et al. Assessment of cortico-spinal tract impairment in multiple system atrophy using transcranial magnetic stimulation. Clin Neurophysiol. 2007;118(4):815–23. doi: 10.1016/j.clinph.2007.01.004. [DOI] [PubMed] [Google Scholar]
  • 27.Van Der Werf YD, et al. Observations on the cortical silent period in Parkinson’s disease. J Neural Transm Suppl. 2007;(72):155–8. doi: 10.1007/978-3-211-73574-9_19. [DOI] [PubMed] [Google Scholar]
  • 28.Hidding U, et al. MEP latency shift after implantation of deep brain stimulation systems in the subthalamic nucleus in patients with advanced Parkinson’s disease. Mov Disord. 2006;21(9):1471–6. doi: 10.1002/mds.20951. [DOI] [PubMed] [Google Scholar]
  • 29.Cincotta M, et al. Mechanisms underlying mirror movements in Parkinson’s disease: a transcranial magnetic stimulation study. Mov Disord. 2006;21(7):1019–25. doi: 10.1002/mds.20850. [DOI] [PubMed] [Google Scholar]
  • 30.Morgante F, et al. Motor cortex plasticity in Parkinson’s disease and levodopa-induced dyskinesias. Brain. 2006;129(Pt 4):1059–69. doi: 10.1093/brain/awl031. [DOI] [PubMed] [Google Scholar]
  • 31.Chuma T, et al. Motor learning of hands with auditory cue in patients with Parkinson’s disease. J Neural Transm. 2006;113(2):175–85. doi: 10.1007/s00702-005-0314-4. [DOI] [PubMed] [Google Scholar]
  • 32.Kuhn AA, et al. Motor cortex inhibition induced by acoustic stimulation. Exp Brain Res. 2004;158(1):120–4. doi: 10.1007/s00221-004-1883-4. [DOI] [PubMed] [Google Scholar]
  • 33.Tamburin S, et al. Abnormal sensorimotor integration is related to disease severity in Parkinson’s disease: a TMS study. Mov Disord. 2003;18(11):1316–24. doi: 10.1002/mds.10515. [DOI] [PubMed] [Google Scholar]
  • 34.Sailer A, et al. Short and long latency afferent inhibition in Parkinson’s disease. Brain. 2003;126(Pt 8):1883–94. doi: 10.1093/brain/awg183. [DOI] [PubMed] [Google Scholar]
  • 35.Bhatia M, Johri S, Behari M. Increased cortical excitability with longer duration of Parkinson’s disease as evaluated by transcranial magnetic stimulation. Neurol India. 2003;51(1):13–5. [PubMed] [Google Scholar]
  • 36.Tamburin S, et al. Abnormal somatotopic arrangement of sensorimotor interactions in dystonic patients. Brain. 2002;125(Pt 12):2719–30. doi: 10.1093/brain/awf279. [DOI] [PubMed] [Google Scholar]
  • 37.Tremblay F, Tremblay LE. Cortico-motor excitability of the lower limb motor representation: a comparative study in Parkinson’s disease and healthy controls. Clin Neurophysiol. 2002;113(12):2006–12. doi: 10.1016/s1388-2457(02)00301-2. [DOI] [PubMed] [Google Scholar]
  • 38.Di Lazzaro V, et al. Direct demonstration of long latency cortico-cortical inhibition in normal subjects and in a patient with vascular parkinsonism. Clin Neurophysiol. 2002;113(11):1673–9. doi: 10.1016/s1388-2457(02)00264-x. [DOI] [PubMed] [Google Scholar]
  • 39.Morita H, et al. Abnormal conditioning effect of transcranial magnetic stimulation on soleus H-reflex during voluntary movement in Parkinson’s disease. Clin Neurophysiol. 2002;113(8):1316–24. doi: 10.1016/s1388-2457(02)00188-8. [DOI] [PubMed] [Google Scholar]
  • 40.Cunic D, et al. Effects of subthalamic nucleus stimulation on motor cortex excitability in Parkinson’s disease. Neurology. 2002;58(11):1665–72. doi: 10.1212/wnl.58.11.1665. [DOI] [PubMed] [Google Scholar]
  • 41.Filippi MM, et al. Effects of motor imagery on motor cortical output topography in Parkinson’s disease. Neurology. 2001;57(1):55–61. doi: 10.1212/wnl.57.1.55. [DOI] [PubMed] [Google Scholar]
  • 42.Kleine BU, et al. Impaired motor cortical inhibition in Parkinson’s disease: motor unit responses to transcranial magnetic stimulation. Exp Brain Res. 2001;138(4):477–83. doi: 10.1007/s002210100731. [DOI] [PubMed] [Google Scholar]
  • 43.Young MS, et al. Stereotactic pallidotomy lengthens the transcranial magnetic cortical stimulation silent period in Parkinson’s disease. Neurology. 1997;49(5):1278–83. doi: 10.1212/wnl.49.5.1278. [DOI] [PubMed] [Google Scholar]
  • 44.Ellaway PH, et al. The relation between bradykinesia and excitability of the motor cortex assessed using transcranial magnetic stimulation in normal and parkinsonian subjects. Electroencephalogr Clin Neurophysiol. 1995;97(3):169–78. doi: 10.1016/0924-980x(94)00336-6. [DOI] [PubMed] [Google Scholar]
  • 45.Abbruzzese G, Marchese R, Trompetto C. Sensory and motor evoked potentials in multiple system atrophy: a comparative study with Parkinson’s disease. Mov Disord. 1997;12(3):315–21. doi: 10.1002/mds.870120309. [DOI] [PubMed] [Google Scholar]
  • 46.Pascual-Leone A, et al. Akinesia in Parkinson’s disease. I. Shortening of simple reaction time with focal, single-pulse transcranial magnetic stimulation. Neurology. 1994;44(5):884–91. doi: 10.1212/wnl.44.5.884. [DOI] [PubMed] [Google Scholar]
  • 47.Rodrigues JP, et al. Spike-timing-related plasticity is preserved in Parkinson’s disease and is enhanced by dopamine: evidence from transcranial magnetic stimulation. Neurosci Lett. 2008;448(1):29–32. doi: 10.1016/j.neulet.2008.10.048. [DOI] [PubMed] [Google Scholar]
  • 48.Bares M, Kanovsky P, Rektor I. Disturbed intracortical excitability in early Parkinson’s disease is l-DOPA dose related: a prospective 12-month paired TMS study. Parkinsonism Relat Disord. 2007;13(8):489–94. doi: 10.1016/j.parkreldis.2007.02.008. [DOI] [PubMed] [Google Scholar]
  • 49.Cantello R, et al. Cortical inhibition in Parkinson’s disease: new insights from early, untreated patients. Neuroscience. 2007;150(1):64–71. doi: 10.1016/j.neuroscience.2007.08.033. [DOI] [PubMed] [Google Scholar]
  • 50.Baumer T, et al. Sensorimotor integration is abnormal in asymptomatic Parkin mutation carriers: a TMS study. Neurology. 2007;69(21):1976–81. doi: 10.1212/01.wnl.0000278109.76607.0a. [DOI] [PubMed] [Google Scholar]
  • 51.Buhmann C, et al. Abnormal excitability of premotor-motor connections in de novo Parkinson’s disease. Brain. 2004;127(Pt 12):2732–46. doi: 10.1093/brain/awh321. [DOI] [PubMed] [Google Scholar]
  • 52.Bares M, et al. Intracortical inhibition and facilitation are impaired in patients with early Parkinson’s disease: a paired TMS study. Eur J Neurol. 2003;10(4):385–9. doi: 10.1046/j.1468-1331.2003.00610.x. [DOI] [PubMed] [Google Scholar]
  • 53.Dauper J, et al. Effects of subthalamic nucleus (STN) stimulation on motor cortex excitability. Neurology. 2002;59(5):700–6. doi: 10.1212/wnl.59.5.700. [DOI] [PubMed] [Google Scholar]
  • 54.Lewis GN, Byblow WD. Altered sensorimotor integration in Parkinson’s disease. Brain. 2002;125(Pt 9):2089–99. doi: 10.1093/brain/awf200. [DOI] [PubMed] [Google Scholar]
  • 55.Pierantozzi M, et al. Effect of apomorphine on cortical inhibition in Parkinson’s disease patients: a transcranial magnetic stimulation study. Exp Brain Res. 2001;141(1):52–62. doi: 10.1007/s002210100839. [DOI] [PubMed] [Google Scholar]
  • 56.Bagnato S, et al. Plasticity of the motor cortex in Parkinson’s disease patients on and off therapy. Mov Disord. 2006;21(5):639–45. doi: 10.1002/mds.20778. [DOI] [PubMed] [Google Scholar]
  • 57.Berardelli A, et al. Cortical inhibition in Parkinson’s disease. A study with paired magnetic stimulation. Brain. 1996;119( Pt 1):71–7. doi: 10.1093/brain/119.1.71. [DOI] [PubMed] [Google Scholar]
  • 58.Chen R, et al. Impairment of motor cortex activation and deactivation in Parkinson’s disease. Clin Neurophysiol. 2001;112(4):600–7. doi: 10.1016/s1388-2457(01)00466-7. [DOI] [PubMed] [Google Scholar]
  • 59.Chu J, et al. Impaired presynaptic inhibition in the motor cortex in Parkinson disease. Neurology. 2009;72(9):842–9. doi: 10.1212/01.wnl.0000343881.27524.e8. [DOI] [PubMed] [Google Scholar]
  • 60.Clouston PD, et al. Apomorphine can increase cutaneous inhibition of motor activity in Parkinson’s disease. Electroencephalogr Clin Neurophysiol. 1996;101(1):8–15. doi: 10.1016/0013-4694(95)00220-0. [DOI] [PubMed] [Google Scholar]
  • 61.Compta Y, et al. The silent period of the thenar muscles to contralateral and ipsilateral deep brain stimulation. Clin Neurophysiol. 2006;117(11):2512–20. doi: 10.1016/j.clinph.2006.08.005. [DOI] [PubMed] [Google Scholar]
  • 62.Cunnington R, et al. Effects of magnetic stimulation over supplementary motor area on movement in Parkinson’s disease. Brain. 1996;119(Pt 3):815–22. doi: 10.1093/brain/119.3.815. [DOI] [PubMed] [Google Scholar]
  • 63.De Rosa A, et al. Neurophysiological evidence of corticospinal tract abnormality in patients with Parkin mutations. J Neurol. 2006;253(3):275–9. doi: 10.1007/s00415-006-0096-0. [DOI] [PubMed] [Google Scholar]
  • 64.Dias AE, et al. Effects of repetitive transcranial magnetic stimulation on voice and speech in Parkinson’s disease. Acta Neurol Scand. 2006;113(2):92–9. doi: 10.1111/j.1600-0404.2005.00558.x. [DOI] [PubMed] [Google Scholar]
  • 65.Dioszeghy P, Hidasi E, Mechler F. Study of central motor functions using magnetic stimulation in Parkinson’s disease. Electromyogr Clin Neurophysiol. 1999;39(2):101–5. [PubMed] [Google Scholar]
  • 66.Fierro B, et al. Motor intracortical inhibition in PD: L-DOPA modulation of high-frequency rTMS effects. Exp Brain Res. 2008;184(4):521–8. doi: 10.1007/s00221-007-1121-y. [DOI] [PubMed] [Google Scholar]
  • 67.Fisher BE, et al. The effect of exercise training in improving motor performance and corticomotor excitability in people with early Parkinson’s disease. Arch Phys Med Rehabil. 2008;89(7):1221–9. doi: 10.1016/j.apmr.2008.01.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Fraix V, et al. Effects of subthalamic nucleus stimulation on motor cortex excitability in Parkinson’s disease. Clin Neurophysiol. 2008;119(11):2513–8. doi: 10.1016/j.clinph.2008.07.217. [DOI] [PubMed] [Google Scholar]
  • 69.Frasson E, et al. Paired transcranial magnetic stimulation for the early diagnosis of corticobasal degeneration. Clin Neurophysiol. 2003;114(2):272–8. doi: 10.1016/s1388-2457(02)00340-1. [DOI] [PubMed] [Google Scholar]
  • 70.Hiraoka K, et al. Premovement facilitation of corticospinal excitability in patients with Parkinson’s disease. Int J Neurosci. 2010;120(2):104–9. doi: 10.3109/00207450903411141. [DOI] [PubMed] [Google Scholar]
  • 71.Hu MT, et al. Limb contractures in levodopa-responsive parkinsonism: a clinical and investigational study of seven new cases. J Neurol. 1999;246(8):671–6. doi: 10.1007/s004150050430. [DOI] [PubMed] [Google Scholar]
  • 72.Ikoma K, Mano Y, Takayanagi T. Pulsed magnetic stimulation and F waves in Parkinson’s disease. Intern Med. 1994;33(2):77–81. doi: 10.2169/internalmedicine.33.77. [DOI] [PubMed] [Google Scholar]
  • 73.Imai T, et al. Reciprocal facilitation of motor evoked potentials immediately before voluntary movements in Parkinson’s disease. Electromyogr Clin Neurophysiol. 1999;39(4):201–6. [PubMed] [Google Scholar]
  • 74.Khedr EM, et al. Lack of post-exercise depression of corticospinal excitability in patients with Parkinson’s disease. Eur J Neurol. 2007;14(7):793–6. doi: 10.1111/j.1468-1331.2007.01858.x. [DOI] [PubMed] [Google Scholar]
  • 75.Leiguarda RC, et al. Limb-kinetic apraxia in corticobasal degeneration: clinical and kinematic features. Mov Disord. 2003;18(1):49–59. doi: 10.1002/mds.10303. [DOI] [PubMed] [Google Scholar]
  • 76.Lou JS, et al. Levodopa normalizes exercise related cortico-motoneuron excitability abnormalities in Parkinson’s disease. Clin Neurophysiol. 2003;114(5):930–7. doi: 10.1016/s1388-2457(03)00040-3. [DOI] [PubMed] [Google Scholar]
  • 77.MacKinnon CD, et al. Pathways mediating abnormal intracortical inhibition in Parkinson’s disease. Ann Neurol. 2005;58(4):516–24. doi: 10.1002/ana.20599. [DOI] [PubMed] [Google Scholar]
  • 78.Manganelli F, et al. Functional involvement of central cholinergic circuits and visual hallucinations in Parkinson’s disease. Brain. 2009;132(Pt 9):2350–5. doi: 10.1093/brain/awp166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Mazzocchio R, et al. Effects of posture-related changes in motor cortical output on central oscillatory activity of pathological origin in humans. Brain Res. 2008;1223:65–72. doi: 10.1016/j.brainres.2008.05.024. [DOI] [PubMed] [Google Scholar]
  • 80.Mir P, et al. Dopaminergic drugs restore facilitatory premotor-motor interactions in Parkinson disease. Neurology. 2005;64(11):1906–12. doi: 10.1212/01.WNL.0000163772.56128.A8. [DOI] [PubMed] [Google Scholar]
  • 81.Nardone R, Lochner P, Tezzon F. Hemiparkinson-hemiatrophy syndrome: a transcranial magnetic stimulation study. Electromyogr Clin Neurophysiol. 2003;43(4):235–40. [PubMed] [Google Scholar]
  • 82.Nardone R, et al. Cholinergic cortical circuits in Parkinson’s disease and in progressive supranuclear palsy: a transcranial magnetic stimulation study. Exp Brain Res. 2005;163(1):128–31. doi: 10.1007/s00221-005-2228-7. [DOI] [PubMed] [Google Scholar]
  • 83.Pierantozzi M, et al. Deep brain stimulation of both subthalamic nucleus and internal globus pallidus restores intracortical inhibition in Parkinson’s disease paralleling apomorphine effects: a paired magnetic stimulation study. Clin Neurophysiol. 2002;113(1):108–13. doi: 10.1016/s1388-2457(01)00694-0. [DOI] [PubMed] [Google Scholar]
  • 84.Sale MV, et al. Pallidotomy does not ameliorate abnormal intracortical inhibition in Parkinson’s disease. J Clin Neurosci. 2010;17(6):711–6. doi: 10.1016/j.jocn.2009.09.038. [DOI] [PubMed] [Google Scholar]
  • 85.Schwingenschuh P, et al. Distinguishing SWEDDs patients with asymmetric resting tremor from Parkinson’s disease: a clinical and electrophysiological study. Mov Disord. 2010;25(5):560–9. doi: 10.1002/mds.23019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Strafella AP, et al. Effects of chronic levodopa and pergolide treatment on cortical excitability in patients with Parkinson’s disease: a transcranial magnetic stimulation study. Clin Neurophysiol. 2000;111(7):1198–202. doi: 10.1016/s1388-2457(00)00316-3. [DOI] [PubMed] [Google Scholar]
  • 87.Thickbroom GW, et al. Motor cortex reorganisation in Parkinson’s disease. J Clin Neurosci. 2006;13(6):639–42. doi: 10.1016/j.jocn.2005.06.013. [DOI] [PubMed] [Google Scholar]
  • 88.Tremblay F, Leonard G, Tremblay L. Corticomotor facilitation associated with observation and imagery of hand actions is impaired in Parkinson’s disease. Exp Brain Res. 2008;185(2):249–57. doi: 10.1007/s00221-007-1150-6. [DOI] [PubMed] [Google Scholar]
  • 89.Ueki Y, et al. Altered plasticity of the human motor cortex in Parkinson’s disease. Ann Neurol. 2006;59(1):60–71. doi: 10.1002/ana.20692. [DOI] [PubMed] [Google Scholar]
  • 90.Vacherot F, et al. Excitability of the lower-limb area of the motor cortex in Parkinson’s disease. Neurophysiol Clin. 2010;40(4):201–8. doi: 10.1016/j.neucli.2010.04.002. [DOI] [PubMed] [Google Scholar]
  • 91.Haug BA, et al. Silent period measurement revives as a valuable diagnostic tool with transcranial magnetic stimulation. Electroencephalogr Clin Neurophysiol. 1992;85(2):158–60. doi: 10.1016/0168-5597(92)90081-l. [DOI] [PubMed] [Google Scholar]
  • 92.Yokota T, Saito Y, Shimizu Y. Increased corticomotoneuronal excitability after peripheral nerve stimulation in dopa-nonresponsive hemiparkinsonism. J Neurol Sci. 1995;129(1):34–9. doi: 10.1016/0022-510x(94)00242-g. [DOI] [PubMed] [Google Scholar]
  • 93.Valls-Sole J, et al. Abnormal facilitation of the response to transcranial magnetic stimulation in patients with Parkinson’s disease. Neurology. 1994;44(4):735–41. doi: 10.1212/wnl.44.4.735. [DOI] [PubMed] [Google Scholar]
  • 94.Priori A, et al. Motor cortical inhibition and the dopaminergic system. Pharmacological changes in the silent period after transcranial brain stimulation in normal subjects, patients with Parkinson’s disease and drug-induced parkinsonism. Brain. 1994;117( Pt 2):317–23. doi: 10.1093/brain/117.2.317. [DOI] [PubMed] [Google Scholar]
  • 95.Pascual-Leone A, et al. Resetting of essential tremor and postural tremor in Parkinson’s disease with transcranial magnetic stimulation. Muscle Nerve. 1994;17(7):800–7. doi: 10.1002/mus.880170716. [DOI] [PubMed] [Google Scholar]
  • 96.Ridding MC, Inzelberg R, Rothwell JC. Changes in excitability of motor cortical circuitry in patients with Parkinson’s disease. Ann Neurol. 1995;37(2):181–8. doi: 10.1002/ana.410370208. [DOI] [PubMed] [Google Scholar]
  • 97.Cincotta M, et al. Motor control in mirror movements: studies with transcranial magnetic stimulation. Suppl Clin Neurophysiol. 2003;56:175–80. doi: 10.1016/s1567-424x(09)70219-3. [DOI] [PubMed] [Google Scholar]
  • 98.Boylan LS, et al. Repetitive transcranial magnetic stimulation to SMA worsens complex movements in Parkinson’s disease. Clin Neurophysiol. 2001;112(2):259–64. doi: 10.1016/s1388-2457(00)00519-8. [DOI] [PubMed] [Google Scholar]
  • 99.Chouinard PA, Paus T. What have We Learned from “Perturbing” the Human Cortical Motor System with Transcranial Magnetic Stimulation? Frontiers in human neuroscience. 2010;4:173. doi: 10.3389/fnhum.2010.00173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Benninger DH, et al. Safety study of 50 Hz repetitive transcranial magnetic stimulation in patients with Parkinson’s disease. Clin Neurophysiol. 2009;120(4):809–15. doi: 10.1016/j.clinph.2009.01.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Huang YZ, et al. Theta burst stimulation of the human motor cortex. Neuron. 2005;45(2):201–6. doi: 10.1016/j.neuron.2004.12.033. [DOI] [PubMed] [Google Scholar]
  • 102.Benninger DH, et al. Intermittent theta-burst transcranial magnetic stimulation for treatment of Parkinson disease. Neurology. 2011;76(7):601–9. doi: 10.1212/WNL.0b013e31820ce6bb. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Suppa A, et al. Lack of LTP-like plasticity in primary motor cortex in Parkinson’s disease. Exp Neurol. 2011;227(2):296–301. doi: 10.1016/j.expneurol.2010.11.020. [DOI] [PubMed] [Google Scholar]
  • 104.Filipovic SR, Rothwell JC, Bhatia K. Slow (1 Hz) repetitive transcranial magnetic stimulation (rTMS) induces a sustained change in cortical excitability in patients with Parkinson’s disease. Clin Neurophysiol. 2010;121(7):1129–37. doi: 10.1016/j.clinph.2010.01.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Gruner U, et al. 1 Hz rTMS preconditioned by tDCS over the primary motor cortex in Parkinson’s disease: effects on bradykinesia of arm and hand. J Neural Transm. 2010;117(2):207–16. doi: 10.1007/s00702-009-0356-0. [DOI] [PubMed] [Google Scholar]
  • 106.Pal E, et al. The impact of left prefrontal repetitive transcranial magnetic stimulation on depression in Parkinson’s disease: a randomized, double-blind, placebo-controlled study. Mov Disord. 2010;25(14):2311–7. doi: 10.1002/mds.23270. [DOI] [PubMed] [Google Scholar]
  • 107.Eggers C, Fink GR, Nowak DA. Theta burst stimulation over the primary motor cortex does not induce cortical plasticity in Parkinson’s disease. J Neurol. 2010;257(10):1669–74. doi: 10.1007/s00415-010-5597-1. [DOI] [PubMed] [Google Scholar]
  • 108.Narayana S, et al. A noninvasive imaging approach to understanding speech changes following deep brain stimulation in Parkinson’s disease. Am J Speech Lang Pathol. 2009;18(2):146–61. doi: 10.1044/1058-0360(2008/08-0004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Baumer T, et al. Effects of DBS, premotor rTMS, and levodopa on motor function and silent period in advanced Parkinson’s disease. Mov Disord. 2009;24(5):672–6. doi: 10.1002/mds.22417. [DOI] [PubMed] [Google Scholar]
  • 110.Sedlackova S, et al. Effect of high frequency repetitive transcranial magnetic stimulation on reaction time, clinical features and cognitive functions in patients with Parkinson’s disease. J Neural Transm. 2009;116(9):1093–101. doi: 10.1007/s00702-009-0259-0. [DOI] [PubMed] [Google Scholar]
  • 111.Koch G, et al. Cerebellar magnetic stimulation decreases levodopa-induced dyskinesias in Parkinson disease. Neurology. 2009;73(2):113–9. doi: 10.1212/WNL.0b013e3181ad5387. [DOI] [PubMed] [Google Scholar]
  • 112.Rothkegel H, et al. Training effects outweigh effects of single-session conventional rTMS and theta burst stimulation in PD patients. Neurorehabil Neural Repair. 2009;23(4):373–81. doi: 10.1177/1545968308322842. [DOI] [PubMed] [Google Scholar]
  • 113.Hamada M, et al. High-frequency rTMS over the supplementary motor area for treatment of Parkinson’s disease. Mov Disord. 2008;23(11):1524–31. doi: 10.1002/mds.22168. [DOI] [PubMed] [Google Scholar]
  • 114.Rektorova I, et al. Dorsolateral prefrontal cortex: a possible target for modulating dyskinesias in Parkinson’s disease by repetitive transcranial magnetic stimulation. Int J Biomed Imaging. 2008;2008:372125. doi: 10.1155/2008/372125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Epstein CM, et al. An open study of repetitive transcranial magnetic stimulation in treatment-resistant depression with Parkinson’s disease. Clin Neurophysiol. 2007;118(10):2189–94. doi: 10.1016/j.clinph.2007.07.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Anninos P, et al. MEG evaluation of Parkinson’s diseased patients after external magnetic stimulation. Acta Neurol Belg. 2007;107(1):5–10. [PubMed] [Google Scholar]
  • 117.Khedr EM, et al. Effect of daily repetitive transcranial magnetic stimulation on motor performance in Parkinson’s disease. Mov Disord. 2006;21(12):2201–5. doi: 10.1002/mds.21089. [DOI] [PubMed] [Google Scholar]
  • 118.Lomarev MP, et al. Placebo-controlled study of rTMS for the treatment of Parkinson’s disease. Mov Disord. 2006;21(3):325–31. doi: 10.1002/mds.20713. [DOI] [PubMed] [Google Scholar]
  • 119.Strafella AP, et al. Corticostriatal functional interactions in Parkinson’s disease: a rTMS/[11C]raclopride PET study. Eur J Neurosci. 2005;22(11):2946–52. doi: 10.1111/j.1460-9568.2005.04476.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Boggio PS, et al. Effect of repetitive TMS and fluoxetine on cognitive function in patients with Parkinson’s disease and concurrent depression. Mov Disord. 2005;20(9):1178–84. doi: 10.1002/mds.20508. [DOI] [PubMed] [Google Scholar]
  • 121.Fregni F, et al. Repetitive transcranial magnetic stimulation is as effective as fluoxetine in the treatment of depression in patients with Parkinson’s disease. J Neurol Neurosurg Psychiatry. 2004;75(8):1171–4. doi: 10.1136/jnnp.2003.027060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Koch G, et al. High-frequency rTMS improves time perception in Parkinson disease. Neurology. 2004;63(12):2405–6. doi: 10.1212/01.wnl.0000147336.19972.82. [DOI] [PubMed] [Google Scholar]
  • 123.Ikeguchi M, et al. Effects of successive repetitive transcranial magnetic stimulation on motor performances and brain perfusion in idiopathic Parkinson’s disease. J Neurol Sci. 2003;209(1–2):41–6. doi: 10.1016/s0022-510x(02)00459-8. [DOI] [PubMed] [Google Scholar]
  • 124.Khedr EM, Farweez HM, Islam H. Therapeutic effect of repetitive transcranial magnetic stimulation on motor function in Parkinson’s disease patients. Eur J Neurol. 2003;10(5):567–72. doi: 10.1046/j.1468-1331.2003.00649.x. [DOI] [PubMed] [Google Scholar]
  • 125.Okabe S, et al. 0.2-Hz repetitive transcranial magnetic stimulation has no add-on effects as compared to a realistic sham stimulation in Parkinson’s disease. Mov Disord. 2003;18(4):382–8. doi: 10.1002/mds.10370. [DOI] [PubMed] [Google Scholar]
  • 126.Gilio F, et al. Repetitive magnetic stimulation of cortical motor areas in Parkinson’s disease: implications for the pathophysiology of cortical function. Mov Disord. 2002;17(3):467–73. doi: 10.1002/mds.1255. [DOI] [PubMed] [Google Scholar]
  • 127.Sommer M, et al. Repetitive paired-pulse transcranial magnetic stimulation affects corticospinal excitability and finger tapping in Parkinson’s disease. Clin Neurophysiol. 2002;113(6):944–50. doi: 10.1016/s1388-2457(02)00061-5. [DOI] [PubMed] [Google Scholar]
  • 128.Shimamoto H, et al. Therapeutic effect and mechanism of repetitive transcranial magnetic stimulation in Parkinson’s disease. J Neurol. 2001;248(Suppl 3):III48–52. doi: 10.1007/pl00007826. [DOI] [PubMed] [Google Scholar]
  • 129.Siebner HR, et al. Short-term motor improvement after sub-threshold 5-Hz repetitive transcranial magnetic stimulation of the primary motor hand area in Parkinson’s disease. J Neurol Sci. 2000;178(2):91–4. doi: 10.1016/s0022-510x(00)00370-1. [DOI] [PubMed] [Google Scholar]
  • 130.Siebner HR, et al. Repetitive transcranial magnetic stimulation causes a short-term increase in the duration of the cortical silent period in patients with Parkinson’s disease. Neurosci Lett. 2000;284(3):147–50. doi: 10.1016/s0304-3940(00)00990-3. [DOI] [PubMed] [Google Scholar]
  • 131.Mally J, Stone TW. Therapeutic and “dose-dependent” effect of repetitive microelectroshock induced by transcranial magnetic stimulation in Parkinson’s disease. J Neurosci Res. 1999;57(6):935–40. [PubMed] [Google Scholar]
  • 132.Mally J, Stone TW. Improvement in Parkinsonian symptoms after repetitive transcranial magnetic stimulation. J Neurol Sci. 1999;162(2):179–84. doi: 10.1016/s0022-510x(98)00318-9. [DOI] [PubMed] [Google Scholar]
  • 133.Ghabra MB, Hallett M, Wassermann EM. Simultaneous repetitive transcranial magnetic stimulation does not speed fine movement in PD. Neurology. 1999;52(4):768–70. doi: 10.1212/wnl.52.4.768. [DOI] [PubMed] [Google Scholar]
  • 134.Siebner HR, et al. Repetitive transcranial magnetic stimulation has a beneficial effect on bradykinesia in Parkinson’s disease. Neuroreport. 1999;10(3):589–94. doi: 10.1097/00001756-199902250-00027. [DOI] [PubMed] [Google Scholar]
  • 135.Pascual-Leone A, et al. Akinesia in Parkinson’s disease. II. Effects of subthreshold repetitive transcranial motor cortex stimulation. Neurology. 1994;44(5):892–8. doi: 10.1212/wnl.44.5.892. [DOI] [PubMed] [Google Scholar]
  • 136.Fregni F, et al. Effects of antidepressant treatment with rTMS and fluoxetine on brain perfusion in PD. Neurology. 2006;66(11):1629–37. doi: 10.1212/01.wnl.0000218194.12054.60. [DOI] [PubMed] [Google Scholar]
  • 137.Arias P, et al. Double-blind, randomized, placebo controlled trial on the effect of 10 days low-frequency rTMS over the vertex on sleep in Parkinson’s disease. Sleep Med. 2010;11(8):759–65. doi: 10.1016/j.sleep.2010.05.003. [DOI] [PubMed] [Google Scholar]
  • 138.Arias P, et al. Controlled trial on the effect of 10 days low-frequency repetitive transcranial magnetic stimulation (rTMS) on motor signs in Parkinson’s disease. Mov Disord. 2010;25(12):1830–8. doi: 10.1002/mds.23055. [DOI] [PubMed] [Google Scholar]
  • 139.Balaz M, et al. The effect of cortical repetitive transcranial magnetic stimulation on cognitive event-related potentials recorded in the subthalamic nucleus. Exp Brain Res. 2010;203(2):317–27. doi: 10.1007/s00221-010-2232-4. [DOI] [PubMed] [Google Scholar]
  • 140.Bornke C, et al. Clinical effects of repetitive transcranial magnetic stimulation versus acute levodopa challenge in Parkinson’s disease. J Neural Transm Suppl. 2004;(68):61–7. doi: 10.1007/978-3-7091-0579-5_7. [DOI] [PubMed] [Google Scholar]
  • 141.Brusa L, et al. Low frequency rTMS of the SMA transiently ameliorates peak-dose LID in Parkinson’s disease. Clin Neurophysiol. 2006;117(9):1917–21. doi: 10.1016/j.clinph.2006.03.033. [DOI] [PubMed] [Google Scholar]
  • 142.Cardoso EF, et al. rTMS treatment for depression in Parkinson’s disease increases BOLD responses in the left prefrontal cortex. Int J Neuropsychopharmacol. 2008;11(2):173–83. doi: 10.1017/S1461145707007961. [DOI] [PubMed] [Google Scholar]
  • 143.Brusa L, et al. Effects of inhibitory rTMS on bladder function in Parkinson’s disease patients. Mov Disord. 2009;24(3):445–8. doi: 10.1002/mds.22434. [DOI] [PubMed] [Google Scholar]
  • 144.del Olmo MF, Bello O, Cudeiro J. Transcranial magnetic stimulation over dorsolateral prefrontal cortex in Parkinson’s disease. Clin Neurophysiol. 2007;118(1):131–9. doi: 10.1016/j.clinph.2006.09.002. [DOI] [PubMed] [Google Scholar]
  • 145.Dragasevic N, et al. Therapeutic efficacy of bilateral prefrontal slow repetitive transcranial magnetic stimulation in depressed patients with Parkinson’s disease: an open study. Mov Disord. 2002;17(3):528–32. doi: 10.1002/mds.10109. [DOI] [PubMed] [Google Scholar]
  • 146.Filipovic SR, et al. Repetitive transcranial magnetic stimulation for levodopa-induced dyskinesias in Parkinson’s disease. Mov Disord. 2009;24(2):246–53. doi: 10.1002/mds.22348. [DOI] [PubMed] [Google Scholar]
  • 147.Furukawa T, et al. Effects of low-frequency repetitive transcranial magnetic stimulation in Parkinson’s disease. Tokai J Exp Clin Med. 2009;34(3):63–71. [PubMed] [Google Scholar]
  • 148.Hartelius L, et al. Short-term effects of repetitive transcranial magnetic stimulation on speech and voice in individuals with Parkinson’s disease. Folia Phoniatr Logop. 2010;62(3):104–9. doi: 10.1159/000287208. [DOI] [PubMed] [Google Scholar]
  • 149.Jacobs JV, et al. The supplementary motor area contributes to the timing of the anticipatory postural adjustment during step initiation in participants with and without Parkinson’s disease. Neuroscience. 2009;164(2):877–85. doi: 10.1016/j.neuroscience.2009.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Kang SY, et al. Characteristics of the sequence effect in Parkinson’s disease. Mov Disord. 2010;25(13):2148–55. doi: 10.1002/mds.23251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Khedr EM, et al. Dopamine levels after repetitive transcranial magnetic stimulation of motor cortex in patients with Parkinson’s disease: preliminary results. Mov Disord. 2007;22(7):1046–50. doi: 10.1002/mds.21460. [DOI] [PubMed] [Google Scholar]
  • 152.Kim JY, et al. Therapeutic effect of repetitive transcranial magnetic stimulation in Parkinson’s disease: analysis of [11C] raclopride PET study. Mov Disord. 2008;23(2):207–11. doi: 10.1002/mds.21787. [DOI] [PubMed] [Google Scholar]
  • 153.Koch G, et al. rTMS of supplementary motor area modulates therapy-induced dyskinesias in Parkinson disease. Neurology. 2005;65(4):623–5. doi: 10.1212/01.wnl.0000172861.36430.95. [DOI] [PubMed] [Google Scholar]
  • 154.Kodama M, et al. Effect of low-frequency repetitive transcranial magnetic stimulation combined with physical therapy on L-dopa-induced painful off-period dystonia in Parkinson’s disease. Am J Phys Med Rehabil. 2011;90(2):150–5. doi: 10.1097/PHM.0b013e3181fc7ccd. [DOI] [PubMed] [Google Scholar]
  • 155.Loscher WN, et al. Abnormal responses to repetitive transcranial magnetic stimulation in multiple system atrophy. Mov Disord. 2007;22(2):174–8. doi: 10.1002/mds.21242. [DOI] [PubMed] [Google Scholar]
  • 156.Mally J, et al. Long-term follow-up study with repetitive transcranial magnetic stimulation (rTMS) in Parkinson’s disease. Brain Res Bull. 2004;64(3):259–63. doi: 10.1016/j.brainresbull.2004.07.004. [DOI] [PubMed] [Google Scholar]
  • 157.Rektorova I, et al. Repetitive transcranial stimulation for freezing of gait in Parkinson’s disease. Mov Disord. 2007;22(10):1518–9. doi: 10.1002/mds.21289. [DOI] [PubMed] [Google Scholar]
  • 158.Sandyk R. Treatment with AC pulsed electromagnetic fields normalizes the latency of the visual evoked response in a multiple sclerosis patient with optic atrophy. Int J Neurosci. 1998;93(3–4):239–50. doi: 10.3109/00207459808986429. [DOI] [PubMed] [Google Scholar]
  • 159.Stephani C, et al. Impairment of motor cortex plasticity in Parkinson’s disease, as revealed by theta-burst-transcranial magnetic stimulation and transcranial random noise stimulation. Parkinsonism Relat Disord. 2011;17(4):297–8. doi: 10.1016/j.parkreldis.2011.01.006. [DOI] [PubMed] [Google Scholar]
  • 160.Suppa A, et al. Dopamine influences primary motor cortex plasticity and dorsal premotor-to-motor connectivity in Parkinson’s disease. Cereb Cortex. 2010;20(9):2224–33. doi: 10.1093/cercor/bhp288. [DOI] [PubMed] [Google Scholar]
  • 161.van Dijk KD, et al. Beneficial effect of transcranial magnetic stimulation on sleep in Parkinson’s disease. Mov Disord. 2009;24(6):878–84. doi: 10.1002/mds.22462. [DOI] [PubMed] [Google Scholar]
  • 162.Filipovic SR, Rothwell JC, Bhatia K. Low-frequency repetitive transcranial magnetic stimulation and off-phase motor symptoms in Parkinson’s disease. J Neurol Sci. 2010;291(1–2):1–4. doi: 10.1016/j.jns.2010.01.017. [DOI] [PubMed] [Google Scholar]
  • 163.Rektor I, Balaz M, Bockova M. Cognitive event-related potentials and oscillations in the subthalamic nucleus. Neurodegener Dis. 2010;7(1–3):160–2. doi: 10.1159/000289228. [DOI] [PubMed] [Google Scholar]
  • 164.Tergau F, et al. Lack of clinical improvement in patients with Parkinson’s disease after low and high frequency repetitive transcranial magnetic stimulation. Electroencephalogr Clin Neurophysiol Suppl. 1999;51:281–8. [PubMed] [Google Scholar]
  • 165.Balaz M, et al. The effect of cortical repetitive transcranial magnetic stimulation on cognitive event-related potentials recorded in the subthalamic nucleus. Experimental brain research. Experimentelle Hirnforschung. Experimentation cerebrale. 2010;203(2):317–27. doi: 10.1007/s00221-010-2232-4. [DOI] [PubMed] [Google Scholar]
  • 166.Stephani C, et al. Impairment of motor cortex plasticity in Parkinson’s disease, as revealed by theta-burst-transcranial magnetic stimulation and transcranial random noise stimulation. Parkinsonism & related disorders. 2011;17(4):297–8. doi: 10.1016/j.parkreldis.2011.01.006. [DOI] [PubMed] [Google Scholar]
  • 167.Eggers C, Fink GR, Nowak DA. Theta burst stimulation over the primary motor cortex does not induce cortical plasticity in Parkinson’s disease. Journal of neurology. 2010;257(10):1669–74. doi: 10.1007/s00415-010-5597-1. [DOI] [PubMed] [Google Scholar]
  • 168.Rothkegel H, et al. Training effects outweigh effects of single-session conventional rTMS and theta burst stimulation in PD patients. Neurorehabilitation and neural repair. 2009;23(4):373–81. doi: 10.1177/1545968308322842. [DOI] [PubMed] [Google Scholar]
  • 169.Kumar R, Chen R, Ashby P. Safety of transcranial magnetic stimulation in patients with implanted deep brain stimulators. Movement disorders: official journal of the Movement Disorder Society. 1999;14(1):157–8. doi: 10.1002/1531-8257(199901)14:1<157::aid-mds1027>3.0.co;2-u. [DOI] [PubMed] [Google Scholar]
  • 170.Pierantozzi M, et al. Deep brain stimulation of both subthalamic nucleus and internal globus pallidus restores intracortical inhibition in Parkinson’s disease paralleling apomorphine effects: a paired magnetic stimulation study. Clinical neurophysiology: official journal of the International Federation of Clinical Neurophysiology. 2002;113(1):108–13. doi: 10.1016/s1388-2457(01)00694-0. [DOI] [PubMed] [Google Scholar]
  • 171.Rothwell J. Transcranial magnetic stimulation as a method for investigating the plasticity of the brain in Parkinson’s disease and dystonia. Parkinsonism & related disorders. 2007;13(Suppl 3):S417–20. doi: 10.1016/S1353-8020(08)70040-3. [DOI] [PubMed] [Google Scholar]
  • 172.Gerloff C, et al. Stimulation over the human supplementary motor area interferes with the organization of future elements in complex motor sequences. Brain. 1997;120( Pt 9):1587–602. doi: 10.1093/brain/120.9.1587. [DOI] [PubMed] [Google Scholar]
  • 173.Gonzalez-Garcia N, et al. Effects of rTMS on Parkinson’s disease: a longitudinal fMRI study. J Neurol. 2011;258(7):1268–80. doi: 10.1007/s00415-011-5923-2. [DOI] [PubMed] [Google Scholar]
  • 174.Borgheresi A, et al. Congenital mirror movements in Parkinson’s disease: clinical and neurophysiological observations. Mov Disord. 2010;25(10):1520–3. doi: 10.1002/mds.23142. [DOI] [PubMed] [Google Scholar]
  • 175.Filipovic SR, et al. Differential effect of linguistic and non-linguistic pen-holding tasks on motor cortex excitability. Exp Brain Res. 2008;191(2):237–46. doi: 10.1007/s00221-008-1517-3. [DOI] [PubMed] [Google Scholar]
  • 176.Kormos TC. Efficacy of rTMS in the treatment of co-morbid anxiety in depressed patients with Parkinson’s disease. Mov Disord. 2007;22(12):1836. doi: 10.1002/mds.21613. [DOI] [PubMed] [Google Scholar]
  • 177.Pascual-Leone A, et al. Induction of visual extinction by rapid-rate transcranial magnetic stimulation of parietal lobe. Neurology. 1994;44(3 Pt 1):494–8. doi: 10.1212/wnl.44.3_part_1.494. [DOI] [PubMed] [Google Scholar]
  • 178.Suppa A, et al. Lack of LTP-like plasticity in primary motor cortex in Parkinson’s disease. Experimental neurology. 2011;227(2):296–301. doi: 10.1016/j.expneurol.2010.11.020. [DOI] [PubMed] [Google Scholar]
  • 179.Kuriakose R, et al. The nature and time course of cortical activation following subthalamic stimulation in Parkinson’s disease. Cerebral cortex. 2010;20(8):1926–36. doi: 10.1093/cercor/bhp269. [DOI] [PubMed] [Google Scholar]
  • 180.Rektor I, Balaz M, Bockova M. Cognitive event-related potentials and oscillations in the subthalamic nucleus. Neuro-degenerative diseases. 2010;7(1–3):160–2. doi: 10.1159/000289228. [DOI] [PubMed] [Google Scholar]
  • 181.Baumer T, et al. Effects of DBS, premotor rTMS, and levodopa on motor function and silent period in advanced Parkinson’s disease. Movement disorders: official journal of the Movement Disorder Society. 2009;24(5):672–6. doi: 10.1002/mds.22417. [DOI] [PubMed] [Google Scholar]
  • 182.Narayana S, et al. A noninvasive imaging approach to understanding speech changes following deep brain stimulation in Parkinson’s disease. American journal of speech-language pathology/American Speech-Language-Hearing Association. 2009;18(2):146–61. doi: 10.1044/1058-0360(2008/08-0004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Gaynor LM, et al. Suppression of beta oscillations in the subthalamic nucleus following cortical stimulation in humans. The European journal of neuroscience. 2008;28(8):1686–95. doi: 10.1111/j.1460-9568.2008.06363.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Fraix V, et al. Effects of subthalamic nucleus stimulation on motor cortex excitability in Parkinson’s disease. Clinical neurophysiology: official journal of the International Federation of Clinical Neurophysiology. 2008;119(11):2513–8. doi: 10.1016/j.clinph.2008.07.217. [DOI] [PubMed] [Google Scholar]
  • 185.Potter-Nerger M, et al. Subthalamic nucleus stimulation restores corticospinal facilitation in Parkinson’s disease. Movement disorders: official journal of the Movement Disorder Society. 2008;23(15):2210–5. doi: 10.1002/mds.22284. [DOI] [PubMed] [Google Scholar]
  • 186.Sailer A, et al. Subthalamic nucleus stimulation modulates afferent inhibition in Parkinson disease. Neurology. 2007;68(5):356–63. doi: 10.1212/01.wnl.0000252812.95774.aa. [DOI] [PubMed] [Google Scholar]
  • 187.Compta Y, et al. The silent period of the thenar muscles to contralateral and ipsilateral deep brain stimulation. Clinical neurophysiology: official journal of the International Federation of Clinical Neurophysiology. 2006;117(11):2512–20. doi: 10.1016/j.clinph.2006.08.005. [DOI] [PubMed] [Google Scholar]
  • 188.Hidding U, et al. MEP latency shift after implantation of deep brain stimulation systems in the subthalamic nucleus in patients with advanced Parkinson’s disease. Movement disorders: official journal of the Movement Disorder Society. 2006;21(9):1471–6. doi: 10.1002/mds.20951. [DOI] [PubMed] [Google Scholar]
  • 189.Kuhn AA, et al. Motor cortex inhibition induced by acoustic stimulation. Experimental brain research. Experimentelle Hirnforschung. Experimentation cerebrale. 2004;158(1):120–4. doi: 10.1007/s00221-004-1883-4. [DOI] [PubMed] [Google Scholar]

RESOURCES