Next Article in Journal
Argon Humidification Exacerbates Antimicrobial and Anti-MRSA kINPen Plasma Activity
Next Article in Special Issue
The Role of V-Set Ig Domain-Containing 4 in Chronic Kidney Disease Models
Previous Article in Journal
Endoscopic Technologies for Peripheral Pulmonary Lesions: From Diagnosis to Therapy
Previous Article in Special Issue
Morphological Evaluation of the Splenic Artery, Its Anatomical Variations and Irrigation Territory
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Application of Repetitive Transcranial Magnetic Stimulation in Neuropathic Pain: A Narrative Review

1
Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital, Bei-Hu Branch, Taipei 10845, Taiwan
2
Department of Physical Medicine and Rehabilitation, National Taiwan University College of Medicine, Taipei 10048, Taiwan
3
Department of Rehabilitation Medicine, First Faculty of Medicine, General University Hospital in Prague, Charles University, 12800 Prague, Czech Republic
4
Physical and Rehabilitation Medicine Unit, Luigi Sacco University Hospital, ASST Fatebenefratelli-Sacco, 20157 Milan, Italy
5
Department of Physical and Rehabilitation Medicine, Hacettepe University Medical School, Ankara 06100, Turkey
6
Department of Physical Medicine and Rehabilitation, West Garden Hospital, Taipei 10864, Taiwan
7
Center for Regional Anesthesia and Pain Medicine, Wang-Fang Hospital, Taipei Medical University, Taipei 11600, Taiwan
*
Authors to whom correspondence should be addressed.
Life 2023, 13(2), 258; https://doi.org/10.3390/life13020258
Submission received: 13 December 2022 / Revised: 8 January 2023 / Accepted: 12 January 2023 / Published: 17 January 2023
(This article belongs to the Special Issue Feature Papers in Medical Research)

Abstract

:
Neuropathic pain, affecting 6.9–10% of the general population, has a negative impact on patients’ quality of life and potentially leads to functional impairment and disability. Repetitive transcranial magnetic stimulation (rTMS)—a safe, indirect and non-invasive technique—has been increasingly applied for treating neuropathic pain. The mechanism underlying rTMS is not yet well understood, and the analgesic effects of rTMS have been inconsistent with respect to different settings/parameters, causing insufficient evidence to determine its efficacy in patients with neuropathic pain. This narrative review aimed to provide an up-to-date overview of rTMS for treating neuropathic pain as well as to summarize the treatment protocols and related adverse effects from existing clinical trials. Current evidence supports the use of 10 Hz HF-rTMS of the primary motor cortex to reduce neuropathic pain, especially in patients with spinal cord injury, diabetic neuropathy and post-herpetic neuralgia. However, the lack of standardized protocols impedes the universal use of rTMS for neuropathic pain. rTMS was hypothesized to achieve analgesic effects by upregulating the pain threshold, inhibiting pain impulse, modulating the brain cortex, altering imbalanced functional connectivity, regulating neurotrophin and increasing endogenous opioid and anti-inflammatory cytokines. Further studies are warranted to explore the differences in the parameters/settings of rTMS for treating neuropathic pain due to different disease types.

1. Introduction

Neuropathic pain has been defined as “pain caused by lesions or diseases of the somatosensory nervous system” by International Association for the Study of Pain [1]. It has a negative impact on patients’ quality of life and potentially leads to functional impairment and disability. Neuropathic pain affects 6.9–10% of the general population [2], 11–57% of people with stroke [3], 53% of people with spinal cord injury (SCI) [4], 26% of people with type 2 diabetes [2] and 21% of people with post-herpes zoster infection [2].
The comprehensive management of neuropathic pain relies on the multidisciplinary approach with a biopsychosocial framework. Conventional pain management includes pharmacotherapy (such as acetaminophen, non-steroidal anti-inflammatory drugs, opioid and neuropathic agents), physical therapy (such as thermotherapy, electrotherapy, hydrotherapy and therapeutic exercises), psychological therapies (such as cognitive behavioral therapy and relaxation techniques), alternative therapies (such as acupuncture, massage and meditation) and injections (such as anesthetics, corticosteroid [5] and dextrose [6]).
Neurostimulation—recently applied in treating neuropathic pain—includes peripheral nerve stimulation [7], dorsal root ganglion stimulation [8], spinal cord stimulation and motor cortex stimulation (MCS) [9], transcranial direct current stimulation (tDCS) [10] and repetitive transcranial magnetic stimulation (rTMS) [11]. Among them, rTMS is a safe, indirect and non-invasive technique that produces a transient magnetic field to induce electrical current for modulating excitability in the brain cortex [12]. It is reported that low-frequency (≦1 Hz) repetitive transcranial magnetic stimulation (LF-rTMS) elicits inhibitory effects [13] and high-frequency (≧5 Hz) repetitive transcranial magnetic stimulation (HF-rTMS) incurs excitatory effects [14]. Based on the aforementioned basic principles, rTMS has been widely used in neuroscientific research and various clinical conditions encompassing depression, anxiety, psychiatric diseases, post-stroke complications (motor deficits, aphasia and dysphagia), movement disorders, consciousness disorders, cognitive impairments and pain [15].
rTMS has been applied for alleviating different pain scenarios, such as neuropathic pain, fibromyalgia and myofascial pain syndrome. An antecedent meta-analysis showed a positive analgesic effect of rTMS for patients with neuropathic pain in comparison with sham treatment [16]. However, the settings of rTMS (such as the intervention site, stimulation frequency, intensity and session) were heterogeneous, and the best protocol for relief of neuropathic pain seems warranted. Therefore, we provide an up-to-date overview of rTMS in treating neuropathic pain and related adverse effects. We summarize the respective treatment protocols for various neuropathic pain types caused by a single disease or lesion from existing clinical trials in order to find out identical parameters to guide clinicians in clinical practice.

2. Literature Search

Studies in the present review were identified by searching two electronic databases, PubMed and Embase, published from the earliest record to 31 October 2022. The key terms included “rTMS OR repetitive transcranial magnetic stimulation” and “neuropathic pain”, with additional restriction to human clinical trials or randomized controlled trials. No language restrictions were applied. Studied were included according to the criteria of (1) clinical trials or randomized controlled trials and (2) specific neuropathic pain caused by a single disease or lesion. Non-relevant studies were excluded by title or abstract (Figure 1). Furthermore, the reference lists of eligible articles were manually searched for additional relevant studies. A relatively small number of studies were enrolled, and selection bias may exist. Indications for rTMS in neuropathic pain are listed below in Table 1.

3. Therapeutic Application of rTMS in Neuropathic Pain

3.1. Central Post-Stroke Pain

Central post-stroke pain is difficult to treat. There are multifactorial etiologies contributing to the development of central post-stroke pain. The parameters of rTMS varied across different studies without reaching a consensus. All these factors complicate the treatment course. Not all patients respond to rTMS targeted on the primary motor cortex [17].
de Oliveira et al. found that applying 10 daily sessions of 10 Hz HF-rTMS to the premotor cortex/dorsolateral prefrontal cortex (DLPFC) at 120% of the resting motor threshold (RMT) in patients with central post-stroke pain did not cause significant analgesic effects compared to sham treatment at 1, 2 and 4 weeks after the last treatment session. Interestingly, variations in the daily visual analog scale (VAS) of pain indicated significant but non-sustained pain reduction right after each stimulation session in 5 out of 10 stimulation days [18]. Ojala et al. compared the analgesic effects of rTMS applied to the primary motor cortex with those of rTMS applied to the secondary somatosensory cortex. All groups showed short-term analgesia following interventions without between-group differences. Furthermore, significant pain reduction at 1-month follow-up was found in the secondary somatosensory cortex group.

3.2. Patients with SCI

Using a stimulation frequency of 5 Hz, Yılmaz et al. applied HF-rTMS to the primary motor cortex in spinal cord injury (SCI) patients with neuropathic pain—without any superior analgesic effects compared to sham rTMS being observed [19]. By applying 5 days of stimulation with 80% RMT stimulation intensity, Kang et al. evaluated the effects of HF-rTMS of the primary motor cortex in SCI patients with neuropathic pain—with insignificant differences in the numeric rating scales of pain compared with sham rTMS being observed [20]. Sun et al. further discovered that in a similar population, extended stimulation duration using a stimulation intensity of 80% RMT for 6 weeks resulted in a greater reduction of pain than sham rTMS [21].

3.3. Phantom Limb Pain

Ahmed et al. applied HF-rTMS to the primary motor cortex in patients with phantom limb pain for five consecutive daily sessions and reported a significant reduction in the VAS of pain at the end of the fifth session (with the effect lasting for 2 months) when compared with the sham group [22]. However, the effect onset was different from that in the study conducted by Malavera et al. [23]. The latter suggested that a significant decrease in pain began on the 15th day after 10 sessions of HF-rTMS, and it could not persist at one month following intervention.

3.4. Radiculopathy

Chronic pain induces neuroplastic changes that lead to alterations in pain circuits, rewriting the resting-state cortical activities and maladaptive brain functional network reorganization [24]. Although lumbosacral radiculopathy results from a peripheral lesion, central sensitization and maladaptive neuroplasticity play an important role in its pathophysiology. A study compared the effectiveness of HF-rTMS, anodal transcranial direct current stimulation (tDCS) and sham stimulation in patients with lumbosacral radiculopathy-related neuropathic pain. The authors found superiority of the analgesic effects pertaining to HF-rTMS applied to the primary motor cortex [25]. Repeated daily sessions of rTMS are believed to expand the analgesic effects. Twice-daily sessions or more pulses per session appeared to be tolerable according to the data of previous studies. Moreover, Schulze et al. found that cumulative use of rTMS rather than a single rTMS stimulation had more therapeutic gain [26].

3.5. Diabetic Neuropathy

A recent study conducted by Yang et al. found that HF-rTMS applied to the hand primary motor cortex in patients with diabetic peripheral neuropathy offered better analgesic effects 1 week post-treatment compared with sham treatment [27]. Onesti et al. advocated a unique H-coil for HF-rTMS of the leg primary motor cortex that was difficult to focus using the conventional coil [28]. A long-lasting analgesic effect up to 3 weeks post-treatment was documented. Although this deep HF-rTMS technique seemingly provided a longer period of pain reduction, patients suffering from chronic refractory diabetic neuropathic pain might need maintenance treatment thereafter.

3.6. Post-Herpetic Neuralgia

In patients with post-herpetic neuralgia, MA et al. [29] and Pei et al. [30] both applied HF-rTMS of the primary motor cortex, with the stimulation frequency of 10 Hz at an intensity of 80% RMT for 10 sessions. They observed significant analgesic effects up to the 3rd post-treatment month. Additionally, Pei et al. compared 10 Hz HF-rTMS, 5 Hz HF-rTMS and sham stimulation in a parallel study design, whereby they demonstrated superior pain reduction and quality-of-life improvement in the 10 Hz HF-rTMS group.

3.7. Neuropathic Orofacial Pain

Lindholm et al. proposed the right secondary somatosensory cortex as a novel target for HF-rTMS in patients with refractory neuropathic orofacial pain [31]. A cross-over study was conducted using one session of active HF-rTMS of the right secondary somatosensory cortex, one session of active HF-rTMS of the right sensorimotor cortex and one sham stimulation. The interval between each stimulation was set at 4 weeks, with the sham intervention being interposed between two active rTMS applications. The study results showed a significantly lower numeric rating scale of pain after stimulation of the right secondary somatosensory cortex.

3.8. Brachial Plexus Injury

Bonifácio de Assis et al. conducted a randomized controlled trial to evaluate the effects of HF-rTMS and tDCS of the primary motor cortex in patients with neuropathic pain after brachial plexus injury [32]. The active group was provided five daily sessions of HF-rTMS and five daily sessions of tDCS with a washout period of 30 days. Each participant underwent 10 active treatment or sham stimulation sessions in both groups. The results revealed a significant reduction in pain in the treatment group, with the effect lasting for at least one month.

4. Parameters/Settings

In our included trials, the frequency of rTMS ranged from 5 to 20 Hz; the intensity ranged between 80% and 120% RMT; and the stimulation pulses ranged from 1000 to 2500 pulses. Until now, there have been no studies using LF-rTMS for managing neuropathic pain. Summarized information from relevant randomized controlled studies, including the characteristics, parameters and outcome, are shown in Table 2.

5. Contraindications to rTMS Application

Given that a magnetic field may induce the unsetting or malfunctioning of implanted devices, the absolute contraindication to rTMS is intracranial metallic hardware, such as brain stimulators [37] or electrodes, epidural cortical stimulators, aneurysm clips [38] or coils, stents and programmable ventriculoperitoneal shunts [39]. Since there are no sufficient safety data regarding rTMS for subjects with implanted electronic devices, the contraindication comprises implantations of the cochlear implant, pacemakers, implantable cardioverter defibrillators and spinal cord stimulators. However, a study reported that rTMS seemed to be safe for implanted electronic devices as long as the internal pulse generator was not close to the rTMS coil [40]. Moreover, it is relatively safe for the application of rTMS in patients with newly implanted electronic devices compatible with magnetic resonance imaging, although further large-scaled studies are warranted for long-term follow-up. Furthermore, rTMS is often contraindicated during pregnancy, based on the concerns for fetus safety. However, currently available evidence reported no detrimental effects on the fetus [41]. The summarized contraindications are listed in Table 3.

6. Adverse Effects of rTMS

Adverse events, such as scalp discomfort, lightheadedness, headache, nausea, tinnitus and hearing loss, are usually mild to moderate. However, severe ones such as seizure and syncope could occasionally happen. Headache has been reported as the most common adverse event (9.7%), followed by scalp discomfort (9.3%) and nausea (5%) [45,47].
In addition, rTMS produces brief but loud coil click sounds, with the peak sound pressure ranging from 110 to 139 dB for the receivers and from 96 to 125 dB for the operators, exceeding 85 dB, i.e., the threshold that potentially causes transient-to-permanent hearing loss [48]. The click sounds during rTMS could significantly increase the auditory threshold and disturb hearing function after stimulation, even under adequate hearing protection in participants with normal hearing. Nevertheless, the elevation of the auditory threshold did not last more than 1 h after a single rTMS session of 20 min [46].
A study reported that the incidence of rTMS-induced seizure was extremely low and usually self-limited [43]. The estimated risk of seizure per session was 0.05%, 0.03%, 0.06% and 0% in those who received HF-rTMS, LF-rTMS, intermittent theta burst rTMS and continuous theta burst rTMS, respectively [45]. No seizure was reported when rTMS was delivered in accordance with the published guidelines and to individuals without seizure risk [45], e.g., brain tumor, severe head trauma, concussion, neurologic disease or medications that lower the seizure threshold [43]. The summarized adverse events of rTMS are listed in Table 3.

7. Mechanism Underlying rTMS in Neuropathic Pain

The analgesic effects of rTMS vary using different parameters. Moreover, the mechanism underlying rTMS is not yet well understood. Therefore, focusing on neuropathic pain, we tried to propose the possible mechanisms regarding HF- and LF-rTMS (Figure 2).
Abbreviations: BDNF, brain-derived neurotrophic factor; DLPFC, dorsolateral prefrontal cortex; HF-rTMS, high-frequency repetitive magnetic stimulation; IL-10, interleukin-10; NMDA, N-methyl-D-aspartate; LF-rTMS, low-frequency repetitive magnetic stimulation; PFC, prefrontal cortex; PMC, premotor cortex; TNF-α, tumor necrosis factor-alpha.

7.1. HF-rTMS

The analgesic effect of HF-rTMS of the primary motor cortex was hypothesized to be similar to that of motor cortex stimulation in modulating pain perception and affective–emotional perception. The proposed mechanisms include (1) upregulating the pain threshold via the pathways from the posterior insula and orbitofrontal cortex to the posterior thalamus [49], (2) modulating emotion and mood via pathways from the posterior insula to the caudal anterior cingulate cortex [49] and (3) inhibiting descending pain impulses via pathway from periaqueductal gray to rostroventromedial medulla [50]. The proposed mechanisms resulted in pain reduction of deafferentation pain caused by damage to pain pathways or maladaptive plasticity [51].
In SCI patients, the cortical excitability and analgesic effects of HF-rTMS applied to the motor cortex responsible for hands and legs have been investigated [34]. The use of rTMS reduces pain regardless of the changes in cortical excitability, implying that the analgesic effect might be independent from the excitable status of the motor cortex [34]. In addition, neuro-navigation is a technique designed to localize intracranial structures precisely using a set of brain images [52]. Non-navigated HF-rTMS and neuro-navigated HF-rTMS targeting the primary motor cortex both appeared efficacious, although the latter seemingly produced more extended analgesic effects in patients with focal neuropathic pain [53,54].
HF-rTMS of the primary motor cortex was found to increase the serum beta-endorphin level [22] in patients with phantom limb pain, with a long-lasting analgesic effect. Regarding the role of endogenous opioids in the analgesic effect, HF-rTMS of the primary motor cortex and DLPFC were investigated for pain threshold/intensity followed by naloxone injection. Naloxone is a competitive opioid receptor antagonist and acts as an opioid antidote. Its injection significantly reduced the analgesic effects of the primary motor cortex group, but it did not change the analgesic effects of the DLPFC and sham groups. The findings demonstrated the role of endogenous opioids in rTMS-induced analgesia and suggested different pain-mediated mechanisms in the primary motor cortex and DLPFC territories [55].
Ketamine is known as an N-methyl-d-aspartate (NMDA) receptor antagonist [56]. In another human study, its injection decreased the analgesic effects of HF-rTMS of the primary motor cortex and DLPFC/premotor cortex territories, which was not associated with changes in cortical excitability. This implies that NMDA receptors play a role in the analgesia of rTMS [57]. Interestingly, HF-rTMS was shown to reduce the pain threshold and induce allodynia in rats via NMDA and α-amino-3-hydroxy-5- methyl-4-isoxazolepropionate/kainate receptors [58].

7.2. LF-rTMS

LF-rTMS of the prefrontal cortex was found to increase regional brain-derived neurotrophic factor (BDNF), tumor necrosis factor-alpha (TNF-α) and interleukin-10 (IL-10) in rats [59]. BDNF is considered to be an upstream neuro-mediator to induce long-lasting synaptic potentiation [60] and a biomarker of cortical excitability and neuronal activity. It is also known as a nociceptive mediator to amplify ascending pain impulses, leading to hyperalgesia and spinal central sensitization [61]. BDNF may be region-specific; yet, it is downregulated in the hippocampus and upregulated in spinal dorsal horn in rats during noxious stimuli [61]. Higher BDNF levels in the blood and cerebrospinal fluid were found in patients with chronic pain than in the asymptomatic controls [62]. Finally, IL-10 serves as an anti-inflammatory mediator. This evidence implies that neurotrophin and anti-neuroinflammatory cytokines might play a role in the analgesic effects of LF-rTMS.

8. rTMS for Comorbidities of Neuropathic Pain

Patients suffering from chronic neuropathic pain often have comorbidities, such as depression and mood disorders. The prevalence rates of depression and major depressive disorders among patients with chronic neuropathic pain were 65.6% [63] and 16.5% [64], respectively.
Imbalanced connectivity of the DLPFC was found in patients with depression [65]. HF-rTMS was applied on the left DLPFC to facilitate neuronal activity and to treat depression [66]. The DLPFC is activated in response to noxious stimuli and is associated with pain suppression through the descending pain pathways [67]. Moreover, the activity of the left DLPFC was negatively correlated with pain [68]. A neurophysiological study revealed that conditioned transcranial magnetic stimulation of the frontal cortex modulated the parameters of motor-evoked potentials, suggesting the existence of cortico-cortical connection [69].
A meta-analysis demonstrated that HF-rTMS of the DLPFC induced a short-term analgesic effect [70], while HF-rTMS of the primary motor cortex showed more evident analgesic effects in patients with neuropathic pain [71]. Moreover, a randomized controlled trial concluded that the analgesic effects of HF-rTMS were not associated with the indirect improvement of depression and were not mediated/predicted by comorbid psychiatric disorders [72].

9. Discussion/Future Perspective

The complexity of neuropathic pain and mechanisms behind rTMS are not completely understood. A recent meta-analysis found that neuropathic pain related to SCI, diabetic neuropathy and post-herpetic neuralgia appeared to be effective after HF-rTMS [71], whereas LF-rTMS showed insignificant analgesic effects. However, there is still lack of evidence for applying rTMS in the general population due to heterogeneity and the small number of available clinical studies.
In combination with our included trials and aforementioned studies, 10 Hz HF-rTMS of the primary motor cortex is recommended as the initial setting for the treatment of neuropathic pain. Regarding stimulation intensity, pulses and sessions, identical parameters have not been determined yet. Similarly, a practical algorithm for applying rTMS for chronic pain was proposed by Lefaucheur et al. in 2019 [73]. Clinically relevant evidence did not support somatotopic effects of HF-rTMS on neuropathic pain [74]. HF-rTMS of the primary motor cortex corresponding to the painful site was less effective than that of the area next to the cortical representation of the painful zone [75]. Hence, 10 Hz HF-rTMS at the intensity of 80–90% RMT could be started by stimulating the primary motor cortex of hands in all patients regardless of the pain location/origin. The stimulation side of the primary motor cortex of hands could be contralateral to the painful side for lateralized neuropathic pain or the left brain cortex for diffuse neuropathic pain. After six–seven sessions, the physicians should re-evaluate whether pain reduction could reach more than 30% of the baseline assessment or two points on a 0–10 numeric rating scale. If the patient does not respond well to the initial session, an alternative stimulating target would be the primary motor cortex corresponding to the sensory/motor processing of face and legs. If there is still no response, the last, but not least, target should be shifted to the left DLPFC, using the beam F3 localization method [76] at 100–110% RMT. The secondary somatosensory cortex is located closely to the insular cortex, which is known to have an important role in pain perception. Strong functional connections between the secondary somatosensory cortex and insular cortex had been found during painful stimuli [77]. rTMS induces neuroplastic changes not only at the stimulation site but also in distant locations probably through cortico–cortical connections and subcortical networks. Therefore, rTMS may exert its analgesic effect in multiple locations of neuronal networks. In cases with good responses to the initial protocol, HF-rTMS can be gradually titrated down from twice a week to once per month [78].
Nevertheless, there are several limitations to the present narrative review. Firstly, despite the fact that we defined inclusion and exclusion criteria and systematically conducted a literature search to reduce selection bias, some studies may have been inevitably omitted. Given the small number of included studies in each etiology category, we had difficulty in pointing out solid and strong opinions without too much bias in only one–two studies. Therefore, we only described their study designs and presented the results objectively in Table 1. Secondly, the algorithm of rTMS application was not tailored according to different diseases. Meanwhile, the conclusion regarding the effective parameters of rTMS from existing meta-analyses might be overgeneralized and not be suitable for various causes of neuropathic pain.
Indeed, the duration of the analgesic effects of HF-rTMS varies from hours [34] to days [78] or one month [31] across different diseases and stimulation parameters. A standard protocol, e.g., a minimal number of sessions in the induction phase, and duration of the maintenance phase and interval between sessions, has not been established. As such, future high-quality studies are definitely awaited.

10. Conclusions

rTMS is a potentially effective and safe treatment of neuropathic pain. Current evidence supports the use of 10 Hz HF-rTMS of the primary motor cortex to reduce neuropathic pain, especially in patients with SCI, diabetic neuropathy and post-herpetic neuralgia. However, the lack of standardized protocols impedes the universal use of rTMS in neuropathic pain. rTMS is hypothesized to achieve analgesic effects by upregulating the pain threshold, inhibiting pain impulse, modulating the brain cortex, altering imbalanced functional connectivity, regulating neurotrophin and increasing endogenous opioids and anti-inflammatory cytokines. Further studies are warranted to explore the differences in the parameters/settings of rTMS for neuropathic pain caused by different diseases.

Author Contributions

Conceptualization, Y.-Y.T. and P.-C.H.; methodology, K.-V.C. and D.-S.H.; software, W.-T.W.; validation, K.M., V.R. and L.Ö.; writing—original draft preparation, Y.-Y.T.; writing—review and editing, K.-V.C.; funding acquisition, K.-V.C. All authors have read and agreed to the published version of the manuscript.

Funding

This study was made possible by (1) research funding from Community and Geriatric Medicine Research Center, National Taiwan University Hospital, Bei-Hu Branch, Taipei, Taiwan; (2) Ministry of Science and Technology (MOST; 106-2314-B-002-180-MY3, 109-2314-B-002-114-MY3, and 109-2314-B-002-127); and (3) Taiwan Society of Ultrasound in Medicine.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the main text of the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Colloca, L.; Ludman, T.; Bouhassira, D.; Baron, R.; Dickenson, A.H.; Yarnitsky, D.; Freeman, R.; Truini, A.; Attal, N.; Finnerup, N.B.; et al. Neuropathic pain. Nat. Rev. Dis. Prim. 2017, 3, 17002. [Google Scholar] [CrossRef] [Green Version]
  2. van Hecke, O.; Austin, S.K.; Khan, R.A.; Smith, B.H.; Torrance, N. Neuropathic pain in the general population: A systematic review of epidemiological studies. Pain 2014, 155, 654–662. [Google Scholar] [CrossRef]
  3. Liampas, A.; Velidakis, N.; Georgiou, T.; Vadalouca, A.; Varrassi, G.; Hadjigeorgiou, G.M.; Tsivgoulis, G.; Zis, P. Prevalence and Management Challenges in Central Post-Stroke Neuropathic Pain: A Systematic Review and Meta-analysis. Adv. Ther. 2020, 37, 3278–3291. [Google Scholar] [CrossRef] [PubMed]
  4. Burke, D.; Fullen, B.M.; Stokes, D.; Lennon, O. Neuropathic pain prevalence following spinal cord injury: A systematic review and meta-analysis. Eur. J. Pain 2017, 21, 29–44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Hsu, P.C.; Chang, K.V.; Wu, W.T.; Wang, J.C.; Özçakar, L. Effects of Ultrasound-Guided Peritendinous and Intrabursal Corticosteroid Injections on Shoulder Tendon Elasticity: A Post Hoc Analysis of a Randomized Controlled Trial. Arch. Phys. Med. Rehabil. 2021, 102, 905–913. [Google Scholar] [CrossRef] [PubMed]
  6. Lin, C.P.; Chang, K.V.; Huang, Y.K.; Wu, W.T.; Özçakar, L. Regenerative Injections Including 5% Dextrose and Platelet-Rich Plasma for the Treatment of Carpal Tunnel Syndrome: A Systematic Review and Network Meta-Analysis. Pharmaceuticals 2020, 13, 49. [Google Scholar] [CrossRef] [Green Version]
  7. Helm, S.; Shirsat, N.; Calodney, A.; Abd-Elsayed, A.; Kloth, D.; Soin, A.; Shah, S.; Trescot, A. Peripheral Nerve Stimulation for Chronic Pain: A Systematic Review of Effectiveness and Safety. Pain Ther. 2021, 10, 985–1002. [Google Scholar] [CrossRef]
  8. Deer, T.R.; Hunter, C.W.; Mehta, P.; Sayed, D.; Grider, J.S.; Lamer, T.J.; Pope, J.E.; Falowski, S.; Provenzano, D.A.; Esposito, M.F.; et al. A Systematic Literature Review of Dorsal Root Ganglion Neurostimulation for the Treatment of Pain. Pain Med. 2020, 21, 1581–1589. [Google Scholar] [CrossRef]
  9. Mo, J.-J.; Hu, W.-H.; Zhang, C.; Wang, X.; Liu, C.; Zhao, B.-T.; Zhou, J.-J.; Zhang, K. Motor cortex stimulation: A systematic literature-based analysis of effectiveness and case series experience. BMC Neurol. 2019, 19, 48. [Google Scholar] [CrossRef] [Green Version]
  10. Zortea, M.; Ramalho, L.; Alves, R.L.; Alves, C.F.d.S.; Braulio, G.; Torres, I.L.d.S.; Fregni, F.; Caumo, W. Transcranial Direct Current Stimulation to Improve the Dysfunction of Descending Pain Modulatory System Related to Opioids in Chronic Non-cancer Pain: An Integrative Review of Neurobiology and Meta-Analysis. Front. Neurosci. 2019, 13, 1218. [Google Scholar] [CrossRef]
  11. Yang, S.; Chang, M.C. Effect of Repetitive Transcranial Magnetic Stimulation on Pain Management: A Systematic Narrative Review. Front. Neurol. 2020, 11, 114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Klomjai, W.; Katz, R.; Lackmy-Vallée, A. Basic principles of transcranial magnetic stimulation (TMS) and repetitive TMS (rTMS). Ann. Phys. Rehabil. Med. 2015, 58, 208–213. [Google Scholar] [CrossRef] [PubMed]
  13. Chen, R.; Classen, J.; Gerloff, C.; Celnik, P.; Wassermann, E.M.; Hallett, M.; Cohen, L.G. Depression of motor cortex excitability by low-frequency transcranial magnetic stimulation. Neurology 1997, 48, 1398–1403. [Google Scholar] [CrossRef] [PubMed]
  14. Pascual-Leone, A.; Valls-Solé, J.; Wassermann, E.M.; Hallett, M. Responses to rapid-rate transcranial magnetic stimulation of the human motor cortex. Brain 1994, 117 Pt 4, 847–858. [Google Scholar] [CrossRef] [PubMed]
  15. Tomeh, A.; Yusof Khan, A.H.K.; Inche Mat, L.N.; Basri, H.; Wan Sulaiman, W.A. Repetitive Transcranial Magnetic Stimulation of the Primary Motor Cortex beyond Motor Rehabilitation: A Review of the Current Evidence. Brain Sci. 2022, 12, 761. [Google Scholar] [CrossRef] [PubMed]
  16. Leung, A.; Donohue, M.; Xu, R.; Lee, R.; Lefaucheur, J.P.; Khedr, E.M.; Saitoh, Y.; André-Obadia, N.; Rollnik, J.; Wallace, M.; et al. rTMS for suppressing neuropathic pain: A meta-analysis. J. Pain 2009, 10, 1205–1216. [Google Scholar] [CrossRef]
  17. Lefaucheur, J.P.; Drouot, X.; Menard-Lefaucheur, I.; Zerah, F.; Bendib, B.; Cesaro, P.; Keravel, Y.; Nguyen, J.P. Neurogenic pain relief by repetitive transcranial magnetic cortical stimulation depends on the origin and the site of pain. J. Neurol. Neurosurg. Psychiatry 2004, 75, 612–616. [Google Scholar] [CrossRef] [Green Version]
  18. de Oliveira, R.A.; de Andrade, D.C.; Mendonça, M.; Barros, R.; Luvisoto, T.; Myczkowski, M.L.; Marcolin, M.A.; Teixeira, M.J. Repetitive transcranial magnetic stimulation of the left premotor/dorsolateral prefrontal cortex does not have analgesic effect on central poststroke pain. J. Pain 2014, 15, 1271–1281. [Google Scholar] [CrossRef]
  19. Yılmaz, B.; Kesikburun, S.; Yaşar, E.; Tan, A.K. The effect of repetitive transcranial magnetic stimulation on refractory neuropathic pain in spinal cord injury. J. Spinal Cord Med. 2014, 37, 397–400. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Kang, B.S.; Shin, H.I.; Bang, M.S. Effect of repetitive transcranial magnetic stimulation over the hand motor cortical area on central pain after spinal cord injury. Arch. Phys. Med. Rehabil. 2009, 90, 1766–1771. [Google Scholar] [CrossRef]
  21. Sun, X.; Long, H.; Zhao, C.; Duan, Q.; Zhu, H.; Chen, C.; Sun, W.; Ju, F.; Sun, X.; Zhao, Y.; et al. Analgesia-enhancing effects of repetitive transcranial magnetic stimulation on neuropathic pain after spinal cord injury:An fNIRS study. Restor. Neurol. Neurosci. 2019, 37, 497–507. [Google Scholar] [CrossRef] [PubMed]
  22. Ahmed, M.A.; Mohamed, S.A.; Sayed, D. Long-term antalgic effects of repetitive transcranial magnetic stimulation of motor cortex and serum beta-endorphin in patients with phantom pain. Neurol. Res. 2011, 33, 953–958. [Google Scholar] [CrossRef] [PubMed]
  23. Malavera, A.; Silva, F.A.; Fregni, F.; Carrillo, S.; Garcia, R.G. Repetitive Transcranial Magnetic Stimulation for Phantom Limb Pain in Land Mine Victims: A Double-Blinded, Randomized, Sham-Controlled Trial. J. Pain 2016, 17, 911–918. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Seifert, F.; Maihöfner, C. Functional and structural imaging of pain-induced neuroplasticity. Curr. Opin. Anaesthesiol. 2011, 24, 515–523. [Google Scholar] [CrossRef] [PubMed]
  25. Attal, N.; Ayache, S.S.; Ciampi De Andrade, D.; Mhalla, A.; Baudic, S.; Jazat, F.; Ahdab, R.; Neves, D.O.; Sorel, M.; Lefaucheur, J.P.; et al. Repetitive transcranial magnetic stimulation and transcranial direct-current stimulation in neuropathic pain due to radiculopathy: A randomized sham-controlled comparative study. Pain 2016, 157, 1224–1231. [Google Scholar] [CrossRef] [PubMed]
  26. Schulze, L.; Feffer, K.; Lozano, C.; Giacobbe, P.; Daskalakis, Z.J.; Blumberger, D.M.; Downar, J. Number of pulses or number of sessions? An open-label study of trajectories of improvement for once-vs. twice-daily dorsomedial prefrontal rTMS in major depression. Brain Stimul. 2018, 11, 327–336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Yang, S.; Kwak, S.G.; Choi, G.S.; Chang, M.C. Short-term Effect of Repetitive Transcranial Magnetic Stimulation on Diabetic Peripheral Neuropathic Pain. Pain Physician 2022, 25, E203–E209. [Google Scholar]
  28. Onesti, E.; Gabriele, M.; Cambieri, C.; Ceccanti, M.; Raccah, R.; Di Stefano, G.; Biasiotta, A.; Truini, A.; Zangen, A.; Inghilleri, M. H-coil repetitive transcranial magnetic stimulation for pain relief in patients with diabetic neuropathy. Eur. J. Pain 2013, 17, 1347–1356. [Google Scholar] [CrossRef]
  29. Ma, S.M.; Ni, J.X.; Li, X.Y.; Yang, L.Q.; Guo, Y.N.; Tang, Y.Z. High-Frequency Repetitive Transcranial Magnetic Stimulation Reduces Pain in Postherpetic Neuralgia. Pain Med. 2015, 16, 2162–2170. [Google Scholar] [CrossRef] [Green Version]
  30. Pei, Q.; Wu, B.; Tang, Y.; Yang, X.; Song, L.; Wang, N.; Li, Y.; Sun, C.; Ma, S.; Ni, J. Repetitive Transcranial Magnetic Stimulation at Different Frequencies for Postherpetic Neuralgia: A Double-Blind, Sham-Controlled, Randomized Trial. Pain Physician 2019, 22, E303–E313. [Google Scholar] [PubMed]
  31. Lindholm, P.; Lamusuo, S.; Taiminen, T.; Pesonen, U.; Lahti, A.; Virtanen, A.; Forssell, H.; Hietala, J.; Hagelberg, N.; Pertovaara, A.; et al. Right secondary somatosensory cortex-a promising novel target for the treatment of drug-resistant neuropathic orofacial pain with repetitive transcranial magnetic stimulation. Pain 2015, 156, 1276–1283. [Google Scholar] [CrossRef]
  32. Bonifácio de Assis, E.D.; Martins, W.K.N.; de Carvalho, C.D.; Ferreira, C.M.; Gomes, R.; de Almeida Rodrigues, E.T.; Meira, U.M.; de Holanda, L.J.; Lindquist, A.R.; Morya, E.; et al. Effects of rTMS and tDCS on neuropathic pain after brachial plexus injury: A randomized placebo-controlled pilot study. Sci. Rep. 2022, 12, 1440. [Google Scholar] [CrossRef] [PubMed]
  33. Ojala, J.; Vanhanen, J.; Harno, H.; Lioumis, P.; Vaalto, S.; Kaunisto, M.A.; Putaala, J.; Kangasniemi, M.; Kirveskari, E.; Mäkelä, J.P.; et al. A Randomized, Sham-Controlled Trial of Repetitive Transcranial Magnetic Stimulation Targeting M1 and S2 in Central Poststroke Pain: A Pilot Trial. Neuromodulation 2022, 25, 538–548. [Google Scholar] [CrossRef] [PubMed]
  34. Jetté, F.; Côté, I.; Meziane, H.B.; Mercier, C. Effect of single-session repetitive transcranial magnetic stimulation applied over the hand versus leg motor area on pain after spinal cord injury. Neurorehabil. Neural Repair 2013, 27, 636–643. [Google Scholar] [CrossRef]
  35. Nardone, R.; Höller, Y.; Langthaler, P.B.; Lochner, P.; Golaszewski, S.; Schwenker, K.; Brigo, F.; Trinka, E. rTMS of the prefrontal cortex has analgesic effects on neuropathic pain in subjects with spinal cord injury. Spinal Cord 2017, 55, 20–25. [Google Scholar] [CrossRef] [PubMed]
  36. Zhao, C.G.; Sun, W.; Ju, F.; Wang, H.; Sun, X.L.; Mou, X.; Yuan, H. Analgesic Effects of Directed Repetitive Transcranial Magnetic Stimulation in Acute Neuropathic Pain After Spinal Cord Injury. Pain Med. 2020, 21, 1216–1223. [Google Scholar] [CrossRef]
  37. Deng, Z.D.; Lisanby, S.H.; Peterchev, A.V. Transcranial magnetic stimulation in the presence of deep brain stimulation implants: Induced electrode currents. Annu. Int. Conf. IEEE Eng. Med. Biol. Soc. 2010, 2010, 6821–6824. [Google Scholar] [CrossRef] [PubMed]
  38. Hsieh, T.H.; Dhamne, S.C.; Chen, J.J.; Carpenter, L.L.; Anastasio, E.M.; Pascual-Leone, A.; Rotenberg, A. Minimal heating of aneurysm clips during repetitive transcranial magnetic stimulation. Clin. Neurophysiol. 2012, 123, 1471–1473. [Google Scholar] [CrossRef] [Green Version]
  39. Lefranc, M.; Ko, J.Y.; Peltier, J.; Fichten, A.; Desenclos, C.; Macron, J.M.; Toussaint, P.; Le Gars, D.; Petitjean, M. Effect of transcranial magnetic stimulation on four types of pressure-programmable valves. Acta Neurochir. 2010, 152, 689–697. [Google Scholar] [CrossRef]
  40. Reveles Jensen, K.H.; Navntoft, C.A.; Sindahl, C.H.; Cayé-Thomasen, P.; Jørgensen, M.B. Cochlear implant should not be absolute contraindication for electroconvulsive therapy and transcranial magnetic stimulation. Brain Stimul. 2020, 13, 1464–1466. [Google Scholar] [CrossRef]
  41. Pridmore, S.; Turnier-Shea, Y.; Rybak, M.; Pridmore, W. Transcranial Magnetic Stimulation (TMS) during pregnancy: A fetal risk factor. Australas. Psychiatry 2021, 29, 226–229. [Google Scholar] [CrossRef] [PubMed]
  42. Philip, N.S.; Carpenter, S.L.; Carpenter, L.L. Safe use of repetitive transcranial magnetic stimulation in patients with implanted vagus nerve stimulators. Brain Stimul. 2014, 7, 608–612. [Google Scholar] [CrossRef]
  43. Stultz, D.J.; Osburn, S.; Burns, T.; Pawlowska-Wajswol, S.; Walton, R. Transcranial Magnetic Stimulation (TMS) Safety with Respect to Seizures: A Literature Review. Neuropsychiatr. Dis. Treat 2020, 16, 2989–3000. [Google Scholar] [CrossRef] [PubMed]
  44. Rossi, S.; Hallett, M.; Rossini, P.M.; Pascual-Leone, A. Safety, ethical considerations, and application guidelines for the use of transcranial magnetic stimulation in clinical practice and research. Clin. Neurophysiol. 2009, 120, 2008–2039. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Lerner, A.J.; Wassermann, E.M.; Tamir, D.I. Seizures from transcranial magnetic stimulation 2012–2016: Results of a survey of active laboratories and clinics. Clin. Neurophysiol. 2019, 130, 1409–1416. [Google Scholar] [CrossRef] [PubMed]
  46. Tringali, S.; Perrot, X.; Collet, L.; Moulin, A. Repetitive transcranial magnetic stimulation: Hearing safety considerations. Brain Stimul. 2012, 5, 354–363. [Google Scholar] [CrossRef]
  47. Overvliet, G.M.; Jansen, R.A.C.; van Balkom, A.; van Campen, D.C.; Oudega, M.L.; van der Werf, Y.D.; van Exel, E.; van den Heuvel, O.A.; Dols, A. Adverse events of repetitive transcranial magnetic stimulation in older adults with depression, a systematic review of the literature. Int. J. Geriatr. Psychiatry 2021, 36, 383–392. [Google Scholar] [CrossRef]
  48. Koponen, L.M.; Goetz, S.M.; Tucci, D.L.; Peterchev, A.V. Sound comparison of seven TMS coils at matched stimulation strength. Brain Stimul. 2020, 13, 873–880. [Google Scholar] [CrossRef]
  49. Kishima, H.; Saitoh, Y.; Osaki, Y.; Nishimura, H.; Kato, A.; Hatazawa, J.; Yoshimine, T. Motor cortex stimulation in patients with deafferentation pain: Activation of the posterior insula and thalamus. J. Neurosurg. 2007, 107, 43–48. [Google Scholar] [CrossRef] [Green Version]
  50. García-Larrea, L.; Peyron, R.; Mertens, P.; Gregoire, M.C.; Lavenne, F.; Le Bars, D.; Convers, P.; Mauguière, F.; Sindou, M.; Laurent, B. Electrical stimulation of motor cortex for pain control: A combined PET-scan and electrophysiological study. Pain 1999, 83, 259–273. [Google Scholar] [CrossRef]
  51. Hanakawa, T. Neural mechanisms underlying deafferentation pain: A hypothesis from a neuroimaging perspective. J. Orthop. Sci. 2012, 17, 331–335. [Google Scholar] [CrossRef]
  52. Ivanov, M.; Ciurea, A.V. Neuronavigation. Principles. Surgical technique. J. Med. Life 2009, 2, 29–35. [Google Scholar]
  53. Ayache, S.S.; Ahdab, R.; Chalah, M.A.; Farhat, W.H.; Mylius, V.; Goujon, C.; Sorel, M.; Lefaucheur, J.P. Analgesic effects of navigated motor cortex rTMS in patients with chronic neuropathic pain. Eur. J. Pain 2016, 20, 1413–1422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Jiang, L.; Ji, Y.; Voulalas, P.J.; Keaser, M.; Xu, S.; Gullapalli, R.P.; Greenspan, J.; Masri, R. Motor cortex stimulation suppresses cortical responses to noxious hindpaw stimulation after spinal cord lesion in rats. Brain Stimul. 2014, 7, 182–189. [Google Scholar] [CrossRef] [Green Version]
  55. de Andrade, D.C.; Mhalla, A.; Adam, F.; Texeira, M.J.; Bouhassira, D. Neuropharmacological basis of rTMS-induced analgesia: The role of endogenous opioids. Pain 2011, 152, 320–326. [Google Scholar] [CrossRef] [PubMed]
  56. Sleigh, J.; Harvey, M.; Voss, L.; Denny, B. Ketamine–More mechanisms of action than just NMDA blockade. Trends Anaesth. Crit. Care 2014, 4, 76–81. [Google Scholar] [CrossRef] [Green Version]
  57. Ciampi de Andrade, D.; Mhalla, A.; Adam, F.; Texeira, M.J.; Bouhassira, D. Repetitive transcranial magnetic stimulation induced analgesia depends onN-methyl-d-aspartate glutamate receptors. PAIN 2014, 155, 598–605. [Google Scholar] [CrossRef] [PubMed]
  58. Ambriz-Tututi, M.; Sánchez-González, V.; Drucker-Colín, R. Transcranial magnetic stimulation reduces nociceptive threshold in rats. J. Neurosci. Res. 2012, 90, 1085–1095. [Google Scholar] [CrossRef]
  59. Toledo, R.S.; Stein, D.J.; Sanches, P.R.S.; da Silva, L.S.; Medeiros, H.R.; Fregni, F.; Caumo, W.; Torres, I.L.S. rTMS induces analgesia and modulates neuroinflammation and neuroplasticity in neuropathic pain model rats. Brain Res. 2021, 1762, 147427. [Google Scholar] [CrossRef]
  60. Fritsch, B.; Reis, J.; Martinowich, K.; Schambra, H.M.; Ji, Y.; Cohen, L.G.; Lu, B. Direct current stimulation promotes BDNF-dependent synaptic plasticity: Potential implications for motor learning. Neuron 2010, 66, 198–204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Duric, V.; McCarson, K.E. Neurokinin-1 (NK-1) receptor and brain-derived neurotrophic factor (BDNF) gene expression is differentially modulated in the rat spinal dorsal horn and hippocampus during inflammatory pain. Mol. Pain 2007, 3, 32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Fischer, M.; Wille, G.; Klien, S.; Shanib, H.; Holle, D.; Gaul, C.; Broessner, G. Brain-derived neurotrophic factor in primary headaches. J. Headache Pain 2012, 13, 469–475. [Google Scholar] [CrossRef] [PubMed]
  63. Radat, F.; Margot-Duclot, A.; Attal, N. Psychiatric co-morbidities in patients with chronic peripheral neuropathic pain: A multicentre cohort study. Eur. J. Pain 2013, 17, 1547–1557. [Google Scholar] [CrossRef] [PubMed]
  64. Cherif, F.; Zouari, H.G.; Cherif, W.; Hadded, M.; Cheour, M.; Damak, R. Depression Prevalence in Neuropathic Pain and Its Impact on the Quality of Life. Pain Res. Manag. 2020, 2020, 7408508. [Google Scholar] [CrossRef] [PubMed]
  65. Kaiser, R.H.; Andrews-Hanna, J.R.; Wager, T.D.; Pizzagalli, D.A. Large-Scale Network Dysfunction in Major Depressive Disorder: A Meta-analysis of Resting-State Functional Connectivity. JAMA Psychiatry 2015, 72, 603–611. [Google Scholar] [CrossRef]
  66. Schutter, D.J. Antidepressant efficacy of high-frequency transcranial magnetic stimulation over the left dorsolateral prefrontal cortex in double-blind sham-controlled designs: A meta-analysis. Psychol. Med. 2009, 39, 65–75. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Seminowicz, D.A.; Moayedi, M. The Dorsolateral Prefrontal Cortex in Acute and Chronic Pain. J. Pain 2017, 18, 1027–1035. [Google Scholar] [CrossRef]
  68. Lorenz, J.; Minoshima, S.; Casey, K.L. Keeping pain out of mind: The role of the dorsolateral prefrontal cortex in pain modulation. Brain 2003, 126, 1079–1091. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Civardi, C.; Cantello, R.; Asselman, P.; Rothwell, J.C. Transcranial magnetic stimulation can be used to test connections to primary motor areas from frontal and medial cortex in humans. Neuroimage 2001, 14, 1444–1453. [Google Scholar] [CrossRef]
  70. Che, X.; Cash, R.F.H.; Luo, X.; Luo, H.; Lu, X.; Xu, F.; Zang, Y.F.; Fitzgerald, P.B.; Fitzgibbon, B.M. High-frequency rTMS over the dorsolateral prefrontal cortex on chronic and provoked pain: A systematic review and meta-analysis. Brain Stimul. 2021, 14, 1135–1146. [Google Scholar] [CrossRef]
  71. Jiang, X.; Yan, W.; Wan, R.; Lin, Y.; Zhu, X.; Song, G.; Zheng, K.; Wang, Y.; Wang, X. Effects of repetitive transcranial magnetic stimulation on neuropathic pain: A systematic review and meta-analysis. Neurosci. Biobehav. Rev. 2022, 132, 130–141. [Google Scholar] [CrossRef] [PubMed]
  72. Lindholm, P.; Lamusuo, S.; Taiminen, T.; Virtanen, A.; Pertovaara, A.; Forssell, H.; Hagelberg, N.; Jääskeläinen, S. The analgesic effect of therapeutic rTMS is not mediated or predicted by comorbid psychiatric or sleep disorders. Medicine 2016, 95, e5231. [Google Scholar] [CrossRef] [PubMed]
  73. Lefaucheur, J.P.; Nguyen, J.P. A practical algorithm for using rTMS to treat patients with chronic pain. Neurophysiol. Clin. 2019, 49, 301–307. [Google Scholar] [CrossRef]
  74. Andre-Obadia, N.; Magnin, M.; Simon, E.; Garcia-Larrea, L. Somatotopic effects of rTMS in neuropathic pain? A comparison between stimulation over hand and face motor areas. Eur. J. Pain 2018, 22, 707–715. [Google Scholar] [CrossRef] [PubMed]
  75. Lefaucheur, J.P.; Hatem, S.; Nineb, A.; Ménard-Lefaucheur, I.; Wendling, S.; Keravel, Y.; Nguyen, J.P. Somatotopic organization of the analgesic effects of motor cortex rTMS in neuropathic pain. Neurology 2006, 67, 1998–2004. [Google Scholar] [CrossRef] [PubMed]
  76. Beam, W.; Borckardt, J.J.; Reeves, S.T.; George, M.S. An efficient and accurate new method for locating the F3 position for prefrontal TMS applications. Brain Stimul. 2009, 2, 50–54. [Google Scholar] [CrossRef] [Green Version]
  77. Peltz, E.; Seifert, F.; DeCol, R.; Dörfler, A.; Schwab, S.; Maihöfner, C. Functional connectivity of the human insular cortex during noxious and innocuous thermal stimulation. Neuroimage 2011, 54, 1324–1335. [Google Scholar] [CrossRef] [PubMed]
  78. Lefaucheur, J.P.; Drouot, X.; Ménard-Lefaucheur, I.; Nguyen, J.P. Neuropathic pain controlled for more than a year by monthly sessions of repetitive transcranial magnetic stimulation of the motor cortex. Neurophysiol. Clin. 2004, 34, 91–95. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Flow diagram for the literature search.
Figure 1. Flow diagram for the literature search.
Life 13 00258 g001
Figure 2. Schematic diagram pertaining to the underlying mechanism of repetitive transcranial magnetic stimulation in alleviating neuropathic pain.
Figure 2. Schematic diagram pertaining to the underlying mechanism of repetitive transcranial magnetic stimulation in alleviating neuropathic pain.
Life 13 00258 g002
Table 1. Indications of rTMS in neuropathic pain.
Table 1. Indications of rTMS in neuropathic pain.
Central neuropathic pain:
Post-stroke;
Spinal cord injury;
Phantom limb pain.
Peripheral neuropathic pain:
Radiculopathy;
Diabetic neuropathy;
Post-herpetic neuralgia;
Neuropathic orofacial pain (e.g., trigeminal neuralgia);
Brachial plexus injury.
Table 2. Characteristics, parameters and results of included randomized controlled studies.
Table 2. Characteristics, parameters and results of included randomized controlled studies.
Author (Year)DiseaseBaseline Characteristics:
Mean Age/Number (Male/Female)
rTMS (n = xx), Control or Sham (n = xx)rTMS SiterTMS FrequencyrTMS IntensityrTMS PulsesrTMS SessionOutcome
de Oliveira et al. (2014) [18]Post-stroke56.3
(l1/10)
11, 10Left PMC/DLPFC 10 Hz120%
RMT
1250Daily with 2-day weekend interval for a total of 10 consecutive sessionsNo differences in pain reduction over 1 month compared to the hand motor cortex group and sham group
Ojala rt al (2022) [33]Post-stroke55.8 (8/9)17, 17S2 contralateral to painful site10 Hz90%
RMT
5050
(train duration, 10 s; intertrain pause, 50 s)
10 sessionsSignificant reduction of weekly pain intensity in S3 group compared with the sham group
Kang et al. (2009) [20]SCI54.8
(6/5)
11, 11FDI motor cortex10 Hz80%
RMT
1000
(train duration, 5 s; intertrain pause, 55 s)
Daily, 5 consecutive sessionsNo differences in average NRS reduction compared to the sham group
Jetté et al. (2013) [34]SCI50
(11/5)
16, 16Hand: FDI motor cortex
Leg: vertex motor cortex
10 HzHand: 90%RMT
Leg: 110%RMT
2000
(train duration, 5 s; intertrain pause, 25 s)
One sessionAbout 10% NRS reduction over the first 49 h compared with the sham group
Yılmaz et al. (2014) [19]SCI38.6
(16/0)
9, 7Vertex motor cortex5 Hz110%
RMT
1500
(train duration, 5 s; intertrain pause, 25 s)
Daily for a total of 10 consecutive sessionsNot superior to the sham group
Nardone et al. (2017) [35]SCI43.1
(9/3)
6, 6PFC/DLPFC: 6 cm anterior to the FDI motor cortex10 Hz120% RMT1250 (train duration, 5 s; intertrain pause, 25 s)5 times per week for 2 weeks for a total of 10 consecutive sessionsSignificant VAS reduction over 1 month compared with the sham group
Sun et al. (2019) [21]SCI37
(15/2)
11, 6Hand motor cortex10 Hz80% RMT1200 (train duration, 1.2 s; intertrain pause, 3 s)Daily with 1-day interval per week for a total of 6 weeksGreater NRS reduction after 2 weeks of rTMS sessions than the sham group
Zhao et al. (2020) [36]SCI41.6
(NA)
24, 24Hand motor cortex10 Hz90% RMT1500 (intertrain pause, 3 s)Daily with 1-day interval per week for a total of 3 weeksSignificant NRS reduction on the 3rd day and 1st week post-rTMS compared with the sham group
Ahmed et al. (2011) [22]Phantom limb pain52.0
(13/140
27Motor cortex corresponding to the stump of painful site20 Hz80%
RMT
NA (train duration, 10 s)Daily, 5 consecutive sessionsSignificant VAS reduction over 2 months compared with the sham group; 55%, 52% and 39% VAS reduction on the day, 1st month and 2nd month post-rTMS, respectively
Malavera et al. (2016) [23]Phantom limb pain33.9
(50/4)
27, 27Contralateral leg motor cortex10 Hz90%
RMT
1200 (train duration, 6 s; intertrain pause, 54 s)Daily, 10 consecutive sessions30.44% greater mean VAS reduction on the 15th day post-rTMS, and no differences on the 30th day post-rTMS, than the sham group
Attal et al. (2016) [25]Lumbosacral radiculopathy52.7
(17/18)
21, 11Thenar motor cortex10 Hz80% RMT3000 (train duration, 10 s; intertrain pause, 20 s)Daily, 3 consecutive sessions30.4% mean pain reduction
Onesti et al. (2013) [28]Diabetic neuropathy70.6
(14/9)
23, 23Leg motor cortex20 Hz100% RMT1500 (intertrain pause, 30 s)Daily, 5 sessionsSignificant VAS reduction over 3 weeks compared with the sham group
Yang et al. (2022) [27]Diabetic peripheral neuropathy 60.4
(11/9)
10, 10Left APB motor cortex10 Hz90%
RMT
1000 (train duration, 5 s; intertrain pause, 55 s)Daily, 5 sessionsSignificant NRS reduction from 6.5 ± 0.9 to 3.6 ± 0.7 1 day post-rTMS; non-significant NRS reduction 1 week post-rTMS compared to the sham group
Ma et al. (2015) [29]PHN66.4
(20/20)
20, 20Motor cortex corresponding to a painful site10 Hz80% RMT1500 (train duration, 5 s; intertrain pause, 3 s)5 times per week for 2 weeks for a total of 10 consecutive sessions16.9% mean VAS reduction over 6 months
Pei et al. (2019) [30]PHN66.2
(30/30)
20, 20, 20Motor cortex corresponding to a painful site10 Hz80%
RMT
10 Hz group: 1500 (train duration, 0.5 s; intertrain pause, 3 s)Daily, 10 sessionsSuperior VAS reduction in 10 Hz group over 3 months compared with 5 Hz and sham groups
Lindholm et al. (2015) [31]Drug-resistant neuropathic orofacial pain57.7
(7/13)
10, 6Right S210 Hz90%
RMT
2500 (train duration, 5 s; intertrain pause, 15 s; 15 min break in the middle of session)One sessionLower NRS of pain over 1 month than S1/M1 or sham groups
Bonifácio de Assis et al. (2022) [32]Traumatic BPI32.8
(20/0)
12, 8Contralateral hand motor cortex10 Hz90%
RMT
2500 (train duration, 10 s; intertrain pause, 17 s)Daily, 5 consecutive daysSuperior continuous pain, paroxysmal pain and anxiety reduction over 1 month compared with the sham group
Abbreviations: APB, abductor pollicis brevis muscle; BPI, brachial plexus injury; FDI, first dorsal interosseous muscle; NA, not available; NRS, numeric rating scale; PHN, post-herpetic neuralgia; PMC/DLPFC, premotor cortex/dorsolateral prefrontal cortex; rTMS, repetitive transcranial magnetic stimulation; RMT, resting motor threshold; S1/M1, sensorimotor cortex; S2, secondary somatosensory cortex; SCI, spinal cord injury; VAS, visual analog scale.
Table 3. Contraindications to and adverse effects of rTMS.
Table 3. Contraindications to and adverse effects of rTMS.
Contraindications to rTMSAdverse Effects of rTMS
  • Absolute
  • Intracranial metallic hardware: brain stimulators [37] or electrodes, epidural cortical stimulators, aneurysm clips [38] or coils, stents and programmable ventriculoperitoneal shunts [39]
  • Relative
  • Implanted electronic device: cochlear implants [40], vagus nerve stimulators [42], pacemakers or implantable cardioverter defibrillators and spinal cord stimulators
  • History of seizure [43], epilepsy and transcranial magnetic stimulation -related seizure
  • History of syncope
  • History of vascular, traumatic, tumoral, infectious, or metabolic brain lesion [44]
  • Drugs that potentially lower seizure threshold
  • Special consideration
  • Pregnancy: currently available evidence reported no detrimental effects on the fetus [41]
  • Seizure (<1%) [43,45]
  • Syncope
  • Scalp discomfort, burn and pain
  • Lightheadedness
  • Headache (most common)
  • Nausea and vomiting
  • Tinnitus and hearing loss [46]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Tsai, Y.-Y.; Wu, W.-T.; Han, D.-S.; Mezian, K.; Ricci, V.; Özçakar, L.; Hsu, P.-C.; Chang, K.-V. Application of Repetitive Transcranial Magnetic Stimulation in Neuropathic Pain: A Narrative Review. Life 2023, 13, 258. https://doi.org/10.3390/life13020258

AMA Style

Tsai Y-Y, Wu W-T, Han D-S, Mezian K, Ricci V, Özçakar L, Hsu P-C, Chang K-V. Application of Repetitive Transcranial Magnetic Stimulation in Neuropathic Pain: A Narrative Review. Life. 2023; 13(2):258. https://doi.org/10.3390/life13020258

Chicago/Turabian Style

Tsai, Yuan-Yuan, Wei-Ting Wu, Der-Sheng Han, Kamal Mezian, Vincenzo Ricci, Levent Özçakar, Po-Cheng Hsu, and Ke-Vin Chang. 2023. "Application of Repetitive Transcranial Magnetic Stimulation in Neuropathic Pain: A Narrative Review" Life 13, no. 2: 258. https://doi.org/10.3390/life13020258

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop