285
Views
19
CrossRef citations to date
0
Altmetric
Review

Clinical utility of brain stimulation modalities following traumatic brain injury: current evidence

, , , , &
Pages 1573-1586 | Published online: 30 Jun 2015

Abstract

Traumatic brain injury (TBI) remains the main cause of disability and a major public health problem worldwide. This review focuses on the neurophysiology of TBI, and the rationale and current state of evidence of clinical application of brain stimulation to promote TBI recovery, particularly on consciousness, cognitive function, motor impairments, and psychiatric conditions. We discuss the mechanisms of different brain stimulation techniques including major noninvasive and invasive stimulations. Thus far, most noninvasive brain stimulation interventions have been nontargeted and focused on the chronic phase of recovery after TBI. In the acute stages, there is limited available evidence of the efficacy and safety of brain stimulation to improve functional outcomes. Comparing the studies across different techniques, transcranial direct current stimulation is the intervention that currently has the higher number of properly designed clinical trials, though total number is still small. We recognize the need for larger studies with target neuroplasticity modulation to fully explore the benefits of brain stimulation to effect TBI recovery during different stages of recovery.

Introduction

Traumatic brain injury (TBI) is one of the leading causes of disabilities and death of young adults. It is estimated that 1.7 million cases occur each year in the United States, in which nearly 80% are treated and released from an emergency department.Citation1 Cognitive impairment and neuropsychiatric disorders are the main disabilities,Citation2Citation4 followed by motor deficits.Citation5 To date, there is no optimal pharmaceutical treatment for acute TBI,Citation6 and brain stimulation techniques appear promising as treatment options to improve neuropsychiatric conditions and motor deficits.Citation7 Our review presents the underlying neuroplasticity mechanisms and maladaptive plasticity involved in stages of recovery of TBI. It focuses on the primary and secondary injury phases. To better understand the mechanism, rationale, and current clinical evidence of noninvasive and invasive brain stimulation, we will provide a comprehensive review on how stimulation techniques modulate brain activity, promote recovery, and prevent further damage after TBI.

The effect of neuroplasticity on TBI

Considerable evidence has shown that the brain has an extensive ability of reorganization after damage. Better understanding of neuroplasticity mechanisms permits more appropriate selection of neuromodulation techniques for the treatment of TBI. Neuroplasticity is defined as an intrinsic property of the human nervous system and occurs in adaptation to environmental stress, physiological changes, and life experiences.Citation8 Neuroplasticity plays a role in neural development, homeostasis,Citation9 and in the dynamic recovery process after injury. In TBI, neuroplasticity can be regarded as an adaptation and reorganization to compensate for the initial insult and to attempt to restore function. We will describe the pathophysiological changes and neuroplasticity in the primary and secondary phases of TBI.

The primary injury phase in TBI

Depending on the mechanism of the trauma, the immediate insult to the brain might be focal (subdural, subarachnoid, or epidural hematoma/hemorrhage/contusion), diffuse (widespread disruption of neuronal circuitry/axonal injury), or mixed (diffuse axonal injury with intracerebral hemorrhage). The initial neuronal injury occurs instantlyCitation10 and oftentimes causes irreversible damage to the central nervous system, due to impairment of neuronal cell functions or cell death.Citation11 Irreversible damage occurs due to the impact of a traumatic event at the origin of acceleration–deceleration shearing, or penetrating injury to the tissues and structures of the brain. Initial shearing of axons and blood vessels can cause intracerebral bleeding, which leads to parenchymal hemorrhage resulting in mass effectCitation10 to the brain tissue. In diffuse axonal injuryCitation10 there is deformation to complete disruption of the axons. This disruption/deformation causes loss of connectivity between different areas of the brain, and can negatively impact neural regeneration, leading to dysfunctional interactions. Thus, even a relatively local lesion can lead to extensive functional damage of other areas of the brain.Citation12

The secondary injury phase in TBI

As a result of an early reduction of cerebral vascular autoregulation and loss of blood–brain barrier integrity, gradual diffuse microvascular damage occurs.Citation13 This diffuse damage increases the risk of ischemic injury and leads to cellular death.Citation11 Other changes include release of neurotransmitters, decreased glucose utilization, lactic acid accumulation, reduced activity of adenosine triphosphate (ATP)-reliant ion pumps, increased release of glutamate, Ca2+-induced depolarization, and excitotoxicity. All of these changes may cause anatomical and functional modifications of synaptic transmission.Citation14 The modulation of the series of actions on a synaptic transmission is an important way to promote brain plasticity.

In the first few weeks after brain injury, brain plasticity and functional recovery involve resolution of edema and inflammation.Citation15 After this initial period, neuroplasticity and remyelination are the most important alterations occurring within the first 3 months after injury.Citation15 It is in the acute and subacute stages that there is greatest potential for modification of neural networks, leading to the formation of new anatomical neural connections.Citation16

Therefore, the improvement of function after TBI needs to be targeted at different points in time. In the acute phase, inhibition of glutamatergic neural activity may reduce neurologic injury.Citation17 In the subacute phase, modulation of gamma aminobutyric acid (GABA)ergic suppression may be crucial to minimize the insult and promote recovery. In the chronic phase, modulation of neuroplasticity is desirable to inhibit maladaptive changes and to promote neural network connections. Ultimately, the final outcome in any stage of injury is to maximize functional recovery. A comprehensive review of the neuroplasticity of TBI can be found in Villamar et al.Citation14 In the following “Methods” section, we will discuss the mechanism, rationale, and current evidence of noninvasive and invasive brain stimulation techniques.

Methods

We searched PubMed (1960–2015), CINAHL (1984–2015), ClinicalKey (2012–2015), EMBASE (1974–2015), and OVID databases (1946–2015). As search term keywords, we used: “Transcranial Magnetic Stimulation (TMS)”, “Transcranial Direct Current Stimulation (tDCS)”, “Transcranial Low-Level Light/Laser Therapy (LLLT)”, “Transcranial Light-Emitting Diode (LED)”, “Deep Brain Stimulation (DBS)”, “Disorders of Consciousness (DOC)”, and “Traumatic Brain Injury (TBI)”. Based on our search, 37 clinical studies were included in this review.

Noninvasive brain stimulation

Noninvasive brain stimulation (NIBS) has the ability to modulate neuron firing. It increases synaptic strength, modulates neurotransmitters and excitotoxicity, and modifies neural network connections, and is therefore a promising therapeutic intervention for TBI. The NIBS methods used to modulate brain plasticity discussed in this article include TMS, tDCS, LLLT, and LED.

TMS

TMS is a NIBS instrument that induces electrical currents via Faraday’s principle of electromagnetic induction. Since its first clinical use in 1985 by Barker et alCitation18 the variety of neuropsychiatric conditions being treated by TMS has increased tremendously.Citation19 The coil placed on the scalp generates a magnetic field that induces a flow of an electric current to neural tissue. This type of stimulation can depolarize/hyperpolarize targeted stimulated areas. For this purpose, there are several protocols of single-pulse and paired-pulse TMS. Thus, TMS may be used as a diagnostic tool to evaluate the integrity of the corticospinal tract, spinal cord, and peripheral nerves.

If TMS is used repetitively, ongoing changes in neuronal excitability can be facilitated or inhibited. Those effects are dependent on stimulation parameters. Low-frequency repetitive TMS (rTMS; 1 Hz) is known to reduce the neural activity in the direct stimulated cortical areas, while high-frequency (>5 Hz) TMS generally increases the neural activity. Repetitive rTMS can modulate the activity of the functionally connected brain regions, reorganizing the neuronal network after injury.Citation20 Theta burst stimulation (TBS) – a mode of patterned rTMS – can modulate cortical excitability.Citation21 This stimulation can be given continuously (cTBS) or intermittently (iTBS). When given continuously, it decreases cortical excitability and given intermittently, it facilitates cortical excitability.

The short effects of TMS on brain activity are partially induced by changes in flow of ionic concentration affecting the synaptic activity in the stimulated area.Citation14 The modulatory effects of TMS can outlast the duration of its application. The after-effect duration is influenced by the magnitude and frequency of stimulation.Citation20 Long-term effects are the result of long-term potentiation (LTP)/long-term depression (LTD), which are mechanisms involved in learning. Therefore improvements in cognitive performance are the result of long lasting changes in synaptic strength induced by cumulative effects of consecutive sessions of rTMS. TMS can also mediate release of glutamate or GABA, which may be the reason for its therapeutic effects.Citation14

Clinical results

Our review of the literature yielded seven clinical studies in which, five studiesCitation21Citation25 are case reports, one is an open label study,Citation26 and one is a cross-sectional survey.Citation27 None of the studies addressed use of TMS in the acute phase. Details are included in .

Table 1 TMS use in TBI

Case reports using TMS addressed neurobehavioral improvements in chronic TBI patients. The aims of these studies were to reduce music hallucinations,Citation24 promote tinnitus relief,Citation25 and decrease depression symptomsCitation22 by using low-frequency rTMS. High-frequency rTMSCitation23 and cTBSCitation21 were used to improve consciousnessCitation23,Citation27 and visuospatial neglect,Citation21 respectively. After the stimulation, the outcomes were reduction of depressive symptoms,Citation22 visuospatial neglect,Citation21 and tinnitus.Citation25 In regards to improvement of consciousnessCitation23 and music hallucinations,Citation24 there were only short-term effects observed.

The number of treatment sessions in these studies varied from 10 to 30 sessions. Targeted areas involved the dorsolateral prefrontal cortex (DLPFC),Citation22,Citation23,Citation26 and the temporalCitation24Citation26 and posterior parietal cortex.Citation21 Only two cases used target neuronavigated rTMS.Citation22,Citation24

The largest TMS study was an open label study with 15 mild TBI patients; however, only 12 patients completed the protocol.Citation26 In this study, patients received 20 sessions of high frequency rTMS (10 Hz) at 110% motor threshold over the left DLPFC. The aim of the study was to alleviate post-concussion syndrome (PCS) symptoms, with positive results observed. Reported side effects included headache and sleep disturbances.Citation26

These studies showed potential benefits of TMS in improving neural conductivity by means of recruitment of neurons, axons, and/or dendritic circuits. Thus far, studies with TMS have included highly variable parameters of stimulation (frequency, number of sessions, treatment duration) and targeted areas. As a consequence of the variability, it is still unclear which TMS protocol is more effective. An important issue that deserves attention is the safety of the method. In the reviewed studies, the side effects were transient and no seizures were reported. A major limitation of these studies is that they were all case reports or case series without sham rTMS to verify the findings.

tDCS

Current modulation of human brain function was first described over 200 years ago,Citation28 and the description was further developed in the animal model in the 1950s and 1960s.Citation29Citation33 tDCS has been used as a NIBS technique, by means of two comparatively large rubber electrodes (25–35 cm2) placed on the scalp. This allows a weak current (1 mA–2 mA) to stream from the anode to the cathode. This stimulation is generally applied for 10–20 minutes. Even though the brain scalp absorbs most of the current, the electrical current that reaches the cerebral cortex has sufficient intensity to modify the resting membrane potential and to modulate the activity level of spontaneous excitatory neurons. Therefore, tDCS is regarded as a neuromodulatory NIBS technique.Citation34

Short-term effects of tDCS may be induced by non-synaptic mechanisms due to neuronal resting membrane depolarization. Such changes may alter the transmembrane proteins and electrolysis-related hydrogen ions.Citation35 It has been reported that a 13-minute, single session of tDCS can lead to a 90-minute period of cortical excitability post-stimulation.Citation36 Consecutive sessions of tDCS can prolong those effects for weeks.Citation37 Long-term effects may be associated with LTP and LTD mechanisms.Citation38 Such long-term effects are dependent on modulation of N-Methyl-D-aspartate (NMDA) receptor activation, as well as neuronal hyperpolarization and depolarization. Previous studiesCitation35Citation37 showed that anodal tDCS increases the excitability of the cerebral cortex, and that cathodal stimulation decreases it. On a behavioral level, anodal tDCS may improve motor task performance, language, and memory. In contrast, cathodal tDCS may also increase performance by decreasing over-activation in an area of maladaptive plasticity.

Clinical results

Due to steady maturation of the technology, relatively low cost, and the ease of use there is increased interest in the potential application of tDCS for treatment of TBI. Our literature review yielded no clinical research on tDCS during the acute phase of TBI. details seven tDCS studies.Citation39Citation45 Six clinical studies were found in the chronic phase of TBI, and there was only one studyCitation39 in the subacute phase. In contrast to TMS studies, most tDCS studies were randomized controlled trials or crossover studies. Outcome measures in most of the studies were changes in consciousness and cognitive performance.Citation40Citation44 The first pilot study was designed to assess whether anodal tDCS applied to left DLPFC could improve attention in patients with chronic TBI compared to sham stimulation.Citation40 Nine patients received anodal tDCS (2 mA for 20 minutes) or sham stimulation (2 mA for 1 minute), in a double-blind, crossover manner with intervals of at least 48 hours.Citation40 It was found that anodal tDCS applied to left DLPFC can significantly shorten reaction times when compared to sham. Two randomized controlled trials have explored whether successive applications of anodal tDCS (15 or 10 sessions of 1 mA for 10 minutes) placed over the left DLPFC would promote changes in attention control and memory track formation in severe TBI.Citation39,Citation41 Those trials revealed no significant improvement in cognitive outcome measures.Citation39,Citation41 However, in one study there were changes in electroencephalography (EEG) recordings associated with an LTP-like mechanism in neural networks, and this method was more likely to be sensitive enough to detect cortical changes than attention/working memory performance.Citation41 One double-blind sham-controlled crossover study provided Class II evidence that short-duration tDCS over the left DLPF cortex transiently improves consciousness as measured by Coma Recovery Scale – Revised (CRS-R) assessment in patients with minimally conscious state (MCS).Citation43

Table 2 tDCS use in TBI

The variance of results of all trials is likely to be related to the differences on number of sessions and timing of application during TBI recovery (chronic vs subacute). In conclusion, the potential application of tDCS as a neuromodulatory tool for blocking or suppressing maladaptive plasticity is still unknown.

In regards to motor function recovery after TBI, we found one study that included chronic TBI participants among stroke patients. All patients received bihemispheric tDCS over M1 paired with standard upper extremity physical therapy (24 sessions of 40 minutes, three times per week). They monitored lasting motor function improvementCitation45 and reported positive results 6 months after tDCS stimulation.

LLLT and transcranial light-emitting diode

LLLT is a NIBS technique used to stimulate biological reactionsCitation46 typically used in the recovery of neuropsychiatric conditions.Citation47,Citation48 LLLT uses low-powered laser light at wavelengths from 632–1,064 nm, ranging from 1–1,000 mW. In acute phase after TBI, a decrease in energy transduction and ATP levels occur due to excessive calcium in the mitochondria within nerve cells impairing the oxidative phosphorylation process. The mechanisms involved in LLLT include the modulation of neurobiological function by improving mitochondrial function, promoting increased ATP and release of nitric oxide locally. This process enhances regional cerebral blood flow and brain oxygen, thereby augmenting metabolic capacity.Citation49,Citation50 Light-modulated cell adhesion and proliferation can be increased or decreased depending on wavelengths used and radiation dose.Citation51 Recently, light-emitting diodes (LEDs) have been used as an alternative light source for LLLT.Citation51,Citation52

Rojas et alCitation53 were the first to record LLLT transcranial tissue response in vivo. They observed brain metabolic and antioxidant beneficial effects measured by increases in cytochrome oxidase expression in neuronal cultures. LLLT- induced up-regulation of cytochrome oxidase in the cortex plays a key role in neuronal physiology, serving as an interface between oxidative energy metabolism and cell survival signaling pathways.Citation54,Citation55 In addition, LLLT partially restores enzyme activity obstructed by potassium cyanide – a cytochrome oxidase inhibitor – reducing neuronal cell death caused by this mitochondrial toxin.Citation52 This enzymatic restoration improves cellular activity of brain tissue that has been damaged by TBI.Citation45 Thereby, transcranial LLLT may become a novel therapy to enhance cognitive performance; emotional functions; and neurological conditionsCitation47,Citation56 linked to mitochondrial dysfunction,Citation47 a ubiquitous finding in brain injury due to TBI.

Animal studies showed benefits in laser phototherapy in damaged TBI cerebral tissue. Those benefits were smaller lesions,Citation57Citation59 improved motor behavior performance,Citation60 increased neurogenesis,Citation56 and changes in biochemical levels.Citation61

Clinical results

To the best of our knowledge, there are only three clinical studiesCitation48,Citation51,Citation62 published using light therapy (LLLT and LED) in patients with TBI. details the manuscripts that evaluated those clinical findings. They were either case reports or open label studies. Nawashiro et alCitation62 studied bilateral transcranial LED irradiation in a patient with persistent vegetative state (VS) following severe TBI. They applied the technique to the forehead of the patient to quantify changes in cerebral blood flow. Single-photon emission computerized tomography (SPECT) analysis showed unilateral increase in cerebral blood flow after 30 minutes of LED therapy applied twice a day. Stimulation on left DLPFC was felt to be responsible for improved akinesia in this patient. Naeser et alCitation51 described two cases of chronic mild TBI. The first case was a patient with chronic attentional problems after 7 years of injury. After 8 weeks of LED treatment applications, there was an improvement of attention. This improvement was observed to gradually decline with interruption of treatment for 2 weeks. The second case was a patient treated after multiple concussions who stopped working due to cognitive dysfunction. After 4 months of LED treatment, the patient reportedly returned to full-time work.

Table 3 LLLT/LED use in TBI

Naeser et alCitation48 examined the effect of two identical LED console units placed over the frontal, parietal, and temporal areas in eleven chronic mild traumatic brain injury patients in an open-protocol study. Their study suggested a reduction in post-traumatic stress symptoms and an improvement in working memory and executive functions after treatment application. Those improvements were still reported at 2-month follow-up.

DBS for TBI

In contrast to noninvasive methods, deep brain stimulation (DBS) is a neurosurgical technique that consists of electrical stimulation through electrodes surgically implanted to subcortical areas. In some neurological conditions, DBS is one of the main procedures in functional neurosurgery.Citation63,Citation64 In patients refractory to drug treatment, DBS is the gold standard for the treatment of motor symptoms of Parkinson’s disease.Citation64,Citation65 This surgery involves the implantation of electrodes through electrical conductors in the basal ganglia in both hemispheres.Citation64,Citation65 The areas usually targeted are the thalamus, subthalamic nuclei, and the globus pallidus. Those areas are subjected to electrical signals that stimulate or inhibit neuronal activity on these nuclei and associated circuitry.Citation65Citation67 The electrodes uses high-frequency stimulation of 70–185 Hz and amplitudes of 0.75–4 V.Citation66Citation70 This technique has greater potential for serious complications and psychiatric and cognitive side effects due to the current spread into brain structures surrounding the electrode. Accordingly to Wolz et alCitation71 the side effects may be due to electrode malposition.

Therefore, in patients with TBI,Citation72,Citation73 clinical application of DBS has been less investigated. This technique has been approved by the US Food and Drug Administration (FDA) for the treatment of disabling symptoms of essential tremor and advanced Parkinson’s disease, and is also approved for dystonia and obsessive compulsive disorder.Citation71 In Europe, in addition to these indications, it is used in epilepsy.Citation72 Research has indicated potential positive outcomes for chronic pain, affective disorders, and a small cohort of patients in minimum state of consciousness.Citation73

Despite application for symptomatic post-traumatic diseases such as tremor,Citation73Citation76 Parkinsonism,Citation77 and hemidystonia,Citation68 there is expectation that the use of DBS might be also beneficial to improve cognitive and consciousness deficits in TBI patients.Citation73Citation77

Clinical results

We found 20 studiesCitation68Citation70,Citation75,Citation77Citation92 testing DBS in chronic TBI patients. details those studies. There were 13 case reports,Citation68Citation70,Citation75,Citation78Citation80,Citation85Citation87,Citation89,Citation91,Citation92 two case series,Citation77,Citation81 and five open label studies.Citation82Citation84,Citation88,Citation90 Tsubokawa et alCitation81 reported significant improvements in a series of eight patients, but the intervention was performed early, within less than a year after TBI. Yamamoto et alCitation82Citation84,Citation88,Citation90 studied series reports of VS and MCSCitation90 caused by various kinds of brain damage. One of these studiesCitation82 described that eight of the 21 patients emerged from the VS and became able to obey verbal commands. The criticism of this study arises from the inclusion of patients 4–8 months following injury during a period of spontaneous recovery. Clinical improvements observed in these studies were based on small series or case reports. There are many variables in which functional and biological aspects warrant further investigation. The precise targets in patients with important anatomical injuries need to be defined before DBS can take a therapeutic role in clinical practice in patients with TBI.

Table 4 DBS use in TBI

Some studies were related to improvement of movement disorders,Citation68Citation70,Citation75,Citation77,Citation79,Citation89,Citation91,Citation92 pain,Citation70,Citation86 and self-mutilation.Citation87 The main targets of those studies were the internal globus pallidus and the ventralis intermedius nucleus. The target for self-mutilation symptoms was the posterior hypothalamus. Some studies reported delayed complications, particularly infarction and infection.Citation68,Citation77,Citation78 Animal studies showed that vagus nerve stimulation, another type of invasive stimulation,Citation93 could improve the prognosis of TBI. Since this technology has not been used in clinical studies, it was not included in this review.

Discussion

We discuss our findings in four separate sections: 1) the “Brain stimulation and biomarkers” section; 2) the “Clinical outcomes and recovery” section; 3) the “Comparison of techniques: which one is better for TBI?” section; and 4) the “Safety” section.

Brain stimulation and biomarkers

There are specific types of biomarkers that assist with finding a prognosis, response to treatment, and extent of TBI. Although their utility is clear, there are limited data regarding their reliability as a clinical tool and what the optimal biomarker is in TBI. We discuss a few biomarkers that are currently being tested.

Commonly tested biomarkers are either proteomic, genetic, or observed changes in brain metabolism.Citation94 Changes in motor-evoked potential via single or paired pulse stimulation and effects of rTMS measured by changes in metabolic activity or cerebral oxygen levels using neuroimaging techniquesCitation95 can be considered neurophysiologic biomarkers.

EEG is another potential biomarker. It provides variation in brain activity during stimulation via tDCS or rTMS. There is a suggestion that changes on EEG frequencies, particularly decrease in delta and increase in alpha, can be a biological marker for response of anodal tDCS reflecting increased cortical activity.Citation39

The technique that has been more studied with biomarkers in TBI is DBS. Unlike NIBS techniques, DBS enables more precise access to target structures. It uses electrophysiological effects on feedback control as a biomarker to establish the timing and intensity of stimulation. In addition to changes in brain signals, functional magnetic resonance imaging (fMRI) has also been used to assess cerebral activity related to post-traumatic Parkinsonism symptoms.Citation96

In summary, EEG and neuroimaging are reliable methods to reflect the effects of brain stimulation and could be suitable biomarkers. These markers indicate correlations between structural lesions, metabolic dysfunction, and cortical activity.

Clinical outcomes and recovery

Numerous studies have implied a relationship between clinical severity measures (eg, the Glasgow Coma Scale [GCS] and duration of post-traumatic amnesia [PTA]) and various types of functional outcome measures at different times after brain injury.Citation97 All protocols in this review addressed the subacute or chronic phase of recovery and used different outcome measures, varying from clinical to functional scores.

While neuroimaging as an assessment tool can provide insights into potential relationships between the GCS, PTA, cognitive function, and outcome after TBI,Citation98 it does increase cost. Only four TMS studies assessed functional recovery assisted with neuroimaging technologies, such as positron emission tomography (PET) and resting fMRI.Citation21,Citation22,Citation24,Citation26 The clinical endpoints in those studies were related to clinical neurobehavioral improvements and also other clinical outcomes, such as transitory reduction of music hallucinations.

tDCS studies measured cognitive function using computerized contrast reaction time taskCitation39 and attention/working memory task.Citation41 They used the JFK Coma Recovery Scale Revised to assess consciousness in persistent VS or MCSCitation41 and monitored improvement of motor function using functional independence measures as a primary outcome.Citation45 Three LLLT/LED studiesCitation48,Citation51,Citation62 addressed improvement of cognition after TBI, but only one study included detailed psychological measurements using the Posttraumatic Stress Disorder Checklist – Civilian; the Beck Depression Inventory – II; and the Visual Analog Scale for pain.

The primary outcomes of DBS studiesCitation23 were level of consciousness and changes in JFK Coma Recovery Scale. The secondary outcomes included neurophysiological evaluation, EEG, and auditory brainstem response. Further studies using comparable and standardized clinical and functional outcomes are warranted to investigate benefits of each brain stimulation technique for different post-traumatic conditions. In fact, some studies, especially those using NIBS, used surrogate cognitive outcomes, such as reaction time in neurophysiological tests, thus making it difficult to determine the clinical utility of these techniques. Given that functional outcomes are associated with more variability and less power, future studies need to test functional outcomes in large sample size studies.

Comparison of techniques: which one is better for TBI?

One important question is which technique is most beneficial for the treatment of TBI. Although data to date do not give enough information to respond this question, a few topics can be explored when comparing techniques: 1) efficacy of these techniques when comparing them; 2) differences of the techniques that may be advantageous for TBI treatment; and 3) safety. There is not enough evidence on efficacy to recommend for or against any of these techniques. Most of the studies are open label or case reports, and the few randomized controlled trials are small and/or used surrogate outcomes. Although the most remarkable clinical improvements have been shown with DBS, comparison is difficult as DBS uses longer protocols of stimulation that may be associated with larger clinical and placebo effects. Therefore, two steps are necessary to determine efficacy of these techniques: 1) development of appropriately designed placebo randomized clinical trials with large sample sizes; and 2) development of randomized clinical trials comparing these techniques.

In terms of differences between the techniques, one point for discussion is the focality. tDCS and LLLT are both nonfocal interventions, while rTMS and DBS are more focal interventions. It is unclear whether the nonfocality of tDCS and LLLT are associated with less effect. It may be argued that less focality in TBI may be beneficial to promote neuroplasticity in a wider area, or that focalization may be achieved when combined with behavioral interventions.

Regarding targeting, for the more focal techniques, there is also the question of what target is most optimal. NIBS methods may be applied over several brain areas involved in neuroplasticity processes. How the target is determined plays an important role during the stimulation. Some studies have stimulated the DLPFC region in order to improve neurobehavioral function, PCS, and depression.Citation22,Citation23,Citation26 With the development of functional imaging techniques, there are more options to achieve this goal. Reviewed rTMS studies applied navigational stimulation before and after the stimulation to achieve the specific targetCitation22,Citation24 using MRI and PET scan. This enabled visualization of the lesion and assessment of response to cortical excitability or connectivity of brain network.Citation21,Citation22,Citation24

DBS alters activity patterns to moderate abnormal brain function related to a specific target. Successful stimulation of the ventralis intermedius nucleus of the thalamus, reduced post-traumatic tremors,Citation98 and DBS targeting the subgenual cingulate cortex were used for the treatment of refractory post-traumatic depression.Citation99Citation101 In this context, development of this field will come with best definition of specific targets for specific behaviors.

The use of neurostimulation strategies and their potential role in recovery of TBI needs to be further developed. Different techniques may be optimized when used in combination, depending on the stage of the recovery and the specific needs of the individual.Citation73 In addition, the use of closed loop systems that can in real time change parameters of stimulation according to the neurophysiological response, may optimize the response to brain stimulation. Finally, the combination of chemical stimulation with drugs and brain stimulation may also result in better clinical outcomes.Citation102

Safety

Considering that TBI is characterized by a chronic hyper-excitability state that increases seizure risk, NIBS, especially rTMS, is regarded as a relative contraindication. In the case where there is a remarkable clinical need, the benefits may outweigh the risks of rTMS, especially when these risks can be minimized. A potential venue to reduce risk would be the use of navigated brain stimulation to ensure safely delivered stimulation to the target area, thereby reducing any adverse effects. In addition, studiesCitation103,Citation104 with low-frequency stimulation have reported antiepileptic effects. The current evidence for application of NIBS recommends exclusion of subjects with a history of seizure, subjects taking medications that lower seizure threshold, or those who have metal implants or brain tumors. DBS, on the other hand, is a controversial modality due to its invasive nature. So far, this stimulation is only used on VS or MCS to regulate arousal. The guidelines of safety for each brain stimulation modality used in TBI needs to be further developed.

Conclusion

This review addresses the clinical utility of brain stimulation modalities to reduce disability and enhance recovery after TBI. Neurostimulation may be applied to a great number of debilitating neurological conditions associated with TBI. For this purpose, brain stimulation techniques may play an important role in inducing neuroplasticity and suppressing pathological disinhibition of circuits implicated in maladaptive networks. Improvements of altered state of consciousness, cognition, and psychiatric and motor function have been the main goals of these therapeutic strategies. Although the mechanisms of neuroplasticity induced by those methods are not fully understood, these instruments have shown great potential for clinical application, significantly changing the current rehabilitation protocols of patients with neurological sequelae post-TBI.

Disclosure

The authors report no conflicts of interest in this work.

References

  • FaulMXuLWaldMMCoronadoVGTraumatic Brain Injury in the United States: Emergency Department Visits, Hospitalizations and Deaths 2002–2006Atlanta, GACenters for Disease Control and Prevention, National Center for Injury Prevention and Control2010
  • PrigatanoGPPersonality disturbances associated with traumatic brain injuryJ Consult Clin Psychol19926033603681619090
  • RiggioSTraumatic brain injury and its neurobehavioral sequelaeNeurol Clin2011291354721172569
  • ArciniegasDBWortzelHSEmotional and behavioral dyscontrol after traumatic brain injuryPsychiatr Clin North Am2014371315324529422
  • WalkerWCPickettTCMotor impairment after severe traumatic brain injury: a longitudinal multicenter studyJ Rehabil Res Dev200744797598218075954
  • CarpenterKLCzosnykaMJallohISystemic, local, and imaging biomarkers of brain injury: more needed, and better use of those already established?Front Neurol201562625741315
  • MouhieddieTHAbu-KheirWTranscranial magnetic stimulation and deep brain stimulation in neuropsychiatric disorders: new dimension of therapeutic markers in psychiatryWangKKWZhangZKobeissyFHBiomarkers of Brain Injury and Neurological Disorders1st edBoca RatonCRC Press2014514565
  • Pascual-LeoneAAmediAFregniFMerabetLBThe plastic human brain cortexAnnu Rev Neurosci20052837740116022601
  • TurrigianoGGNelsonSBHomeostatic plasticity in the developing nervous systemNat Rev Neurosci2004529710714735113
  • MustafaAGAlshboulOAPathophysiology of traumatic brain injuryNeurosciences (Riyadh)201318322223423887212
  • AlgattasHHuangJHTraumatic Brain Injury pathophysiology and treatments: early, intermediate, and late phases post-injuryInt J Mol Sci201415130934124381049
  • PovlishockJTKatzDIUpdate of neuropathology and neurological recovery after traumatic brain injuryJ Head Trauma Rehabil2005201769415668572
  • FrattaloneARLingGSModerate and severe traumatic brain injury: pathophysiology and managementNeurosurg Clin N Am201324330931923809027
  • VillamarMFSantos PortillaAFregniFZafonteRNoninvasive brain stimulation to modulate neuroplasticity in traumatic brain injuryNeuromodulation201215432633822882244
  • FawcettJMolecular control of brain plasticity and repairProg Brain Res200917550150919660677
  • WielochTNikolichKMechanisms of neural plasticity following brain injuryCurr Opin Neurobiol200616325826416713245
  • AndriessenTMJacobsBVosPEClinical characteristics and pathophysiological mechanisms of focal and diffuse traumatic brain injuryJ Cell Mol Med201014102381239220738443
  • BarkerATJalinousRFreestonILNon-invasive magnetic stimulation of human motor cortexLancet198518437110611072860322
  • LefaucheurJPAndré-ObadiaNAntalAEvidence-based guidelines on the therapeutic use of repetitive transcranial magnetic stimulation (rTMS)Clin Neurophysiol2014125112150220625034472
  • Demirtas-TatlidedeAVahabzadeh-HaghAMBernabeuMTormosJMPascual-LeoneANoninvasive brain stimulation in traumatic brain injuryJ Head Trauma Rehabil201227427429221691215
  • BonnìSMastropasquaCBozzaliMCaltagironeCKochGTheta burst stimulation improves visuo-spatial attention in a patient with traumatic brain injuryNeurol Sci201334112053205623532550
  • NielsonDMMcKnightCAPatelRNKalninAJMysiwWJPreliminary guidelines for safe and effective use of repetitive transcranial magnetic stimulation in moderate to severe traumatic brain injuryArch Phys Med Rehabil2015964 SupplS138S14425281871
  • Louise-Bender PapeTRosenowJLewisGRepetitive transcranial magnetic stimulation-associated neurobehavioral gains during coma recoveryBrain Stimul200921223520633400
  • CosentinoGGigliaGPalermoAA case of post-traumatic complex auditory hallucinosis treated with rTMSNeurocase201016326727220104391
  • KreuzerPMLandgrebeMFrankELangguthBRepetitive transcranial magnetic stimulation for the treatment of chronic tinnitus after traumatic brain injury: a case studyJ Head Trauma Rehabil201328538638922688213
  • KoskiLKolivakisTYuCChenJKDelaneySPtitoANoninvasive brain stimulation for persistent post-concussion symptoms in mild traumatic brain injuryJ Neurotrauma2015321384424955920
  • PistoiaFSaccoSCaroleiASaràMCorticomotor facilitation in vegetative state: results of a pilot studyArch Phys Med Rehabil20139481599160623385107
  • BartholowRExperimental investigations into the functions of the human brainAm J Med Sci187466134305313
  • BurnsBDThe production of after-bursts in isolated unanesthetized cerebral cortexJ Physiol1954125342744613212709
  • CreutzfeldtODFrommGHKappHInfluence of transcortical d-c currents on cortical neuronal activityExp Neurol1962543645213882165
  • GoldringSO’LearyJLExperimentally derived correlates between ECG and steady cortical potentialJ Neurophysiol195114427528814851004
  • BindmanLJLippoldOCRedfearnJWThe action of brief polarizing currents on the cerebral cortex of the rat (1) during current flow and (2) in the production of long-lasting after-effectsJ Physiol196417236938214199369
  • PurpuraDPMcMurtryJGIntracellular activities and evoked potential changes during polarization of motor cortexJ Neurophysiol19652816618514244793
  • NitscheMACohenLGWassermannEMTranscranial direct current stimulation: state of the art 2008Brain Stimul20081320622320633386
  • NitscheMAPaulusWExcitability changes induced in the human motor cortex by weak transcranial direct current stimulationJ Physiol2000527Pt 363363910990547
  • NitscheMAPaulusWSustained excitability elevations induced by transcranial DC motor cortex stimulation in humansNeurology200157101899190111723286
  • BoggioPSNunesARigonattiSPNitscheMAPascual-LeoneAFregniFRepeated sessions of noninvasive brain DC stimulation is associated with motor function improvement in stroke patientsRestor Neurol Neurosci200725212312917726271
  • LiebetanzDNitscheMATergauFPaulusWPharmacological approach to the mechanisms of transcranial DC-stimulation-induced after-effects of human motor cortex excitabilityBrain2002125Pt 102238224712244081
  • UlamFSheltonCRichardsLCumulative effects of transcranial direct current stimulation on EEG oscillations and attention/working memory during subacute neurorehabilitation of traumatic brain injuryClin Neurophysiol2015126348649624947595
  • KangEKKimDYPaikNJTranscranial direct current stimulation of the left prefrontal cortex improves attention in patients with traumatic brain injury: a pilot studyJ Rehabil Med201244434635022434324
  • LésniakMPolanowskaKSeniówJCzłonkowskaAEffects of repeated anodal tDCS coupled with cognitive training for patients with severe traumatic brain injury: a pilot randomized controlled trialJ Head Trauma Rehabil2014293E20E2923756431
  • AngelakisELioutaEAndreadisNTranscranial direct current stimulation effects in disorders of consciousnessArch Phys Med Rehabil201495228328924035769
  • ThibautABrunoMALedouxDDemertziALaureysStDCS in patients with disorders of consciousness: sham-controlled randomized double-blind studyNeurology201482131112111824574549
  • NaroACalabròRSRussoMCan transcranial direct current stimulation be useful in differentiating unresponsive wakefulness syndrome from minimally conscious state patients?Restor Neurol Neurosci201533215917625588461
  • MiddletonAFritzSLLiuzzoDMNewman-NorlundRHerterTMUsing clinical and robotic assessment tools to examine the feasibility of pairing tDCS with upper extremity physical therapy in patients with stroke and TBI: a consideration-of-concept pilot studyNeuroRehabilitation201435474175425323084
  • HashmiJTHuangYYOsmaniBZSharmaSKNaeserMAHamblinMRRole of low-level laser therapy in neurorehabilitationPM R2010212 Suppl 2S292S30521172691
  • RojasJCGonzalez-LimaFNeurological and psychological applications of transcranial lasers and LEDsBiochem Pharmacol201386444745723806754
  • NaeserMAZafonteRKrengelMHSignificant improvements in cognitive performance post-transcranial, red/near-infrared light-emitting diode treatments in chronic, mild traumatic brain injury: open-protocol studyJ Neurotrauma201431111008101724568233
  • KaruTIPyatibratLVAfanasyevaNICellular effects of low power laser therapy can be mediated by nitric oxideLasers Surg Med200536430731415739174
  • SchifferFJohnstonALRavichandranCPsychological benefits 2 and 4 weeks after a single treatment with near infrared light to the forehead: a pilot study of 10 patients with major depression and anxietyBehav Brain Funct200954619995444
  • NaeserMASaltmarcheAKrengelMHHamblinMRKnightJAImproved cognitive function after transcranial, light-emitting diode treatments in chronic, traumatic brain injury: two case reportsPhotomed Laser Surg201129535135821182447
  • KaruTIPyatibratLVGene expression under laser and light-emitting diodes radiation for modulation of cell adhesion: Possible applications for biotechnologyIUBMB Life201163974775321796755
  • RojasJCLeeJJohnJMGonzalez-LimaFNeuroprotective effects of near-infrared light in an in vivo model of mitochondrial optic neuropathyJ Neurosci200828135111352119074024
  • RojasJCBrucheyAKGonzalez-LimaFLow-level light therapy improves cortical metabolic capacity and memory retentionJ Alzheimers Dis20123274175222850314
  • LiangHLWhelanHTEellsJTPhotobiomodulation partially rescues visual cortical neurons from cyanide induced apoptosisNeuroscience200613963964916464535
  • BarrettDWGonzalez-LimaFTranscranial infrared laser stimulation produces beneficial cognitive and emotional effects in humansNeuroscience2013230132323200785
  • MoreiraMSVelascoITFerreiraLSEffect of laser phototherapy on wound healing following cerebral ischemia by cryogenic injuryJ Photochem Photobiol B2011105320721522024356
  • OronAOronUStreeterJNear infrared transcranial laser therapy applied at various modes to mice following traumatic brain injury significantly reduces long-term neurological deficitsJ Neurotrauma201229240140722040267
  • XuanWVatanseverFHuangLHamblinMRTranscranial low-level laser therapy enhances learning, memory, and neuroprogenitor cells after traumatic brain injury in miceJ Biomed Opt2014191010800325292167
  • OronAOronUStreeterJLow-level laser therapy applied transcranially to mice following traumatic brain injury significantly reduces long-term neurological deficitsJ Neurotrauma200724465165617439348
  • QuirkBJTorbeyMBuchmannEVermaSWhelanHTNear-infrared photobiomodulation in an animal model of traumatic brain injury: improvements at the behavioral and biochemical levelsPhotomed Laser Surg201230952352922793787
  • NawashiroHWadaKNakaiKSatoSFocal increase in cerebral blood flow after treatment with near-infrared light to the forehead in a patient in a persistent vegetative statePhotomed Laser Surg201230423123322047598
  • LeeDJGurkoffGGIzadiAMedial septal nucleus theta frequency deep brain stimulation improves spatial working memory after traumatic brain injuryJ Neurotrauma201330213113923016534
  • SchuepbachWMRauJKnudsenKEARLYSTIM Study GroupNeurostimulation for Parkinson’s disease with early motor complicationsN Engl J Med201336861062223406026
  • WeaverFMFollettKSternMCSP 468 Study GroupBilateral deep brain stimulation vs best medical therapy for patients with advanced Parkinson disease: a randomized controlled trialJAMA2009301637319126811
  • NardoneRHöllerYLeisSInvasive and non-invasive brain stimulation for treatment of neuropathic pain in patients with spinal cord injury: a reviewJ Spinal Cord Med2014371193124090372
  • MüllerUJVogesJSteinerJDeep brain stimulation of the nucleus accumbens for the treatment of addictionAnn N Y Acad Sci2013128211912823227826
  • KimJPChangWSChangJWThe long-term surgical outcomes of secondary hemidystonia associated with post-traumatic brain injuryActa Neurochir (Wien)2012154582383022367408
  • CarvalhoKSSukulVVBooklandMJKochSAConnollyPJDeep brain stimulation of the globus pallidus suppresses post-traumatic dystonic tremorJ Clin Neurosci201421115315523896546
  • LoherTJHasdemirMGBurgunderJMKraussJKLong-term follow-up study of chronic globus pallidus internus stimulation for posttraumatic hemidystoniaJ Neurosurg200092345746010701534
  • WolzMHauschildJKoyJImmediate effects of deep brain stimulation of the subthalamic nucleus on nonmotor symptoms in Parkinson’s diseaseParkinsonism Relat Disord201218899499722682974
  • CarronRChailletAFilipchukAPasillas-LepineWHammondCClosing the loop of deep brain stimulationFront Syst Neurosci2013711224391555
  • ShinSSDixonCEOkonkwoDORichardsonRMNeurostimulation for traumatic brain injuryJ Neurosurg201412151219123125170668
  • GhaffarpasandFRazmkonAKhaliliHDeep brain stimulation in patients with traumatic brain injury; facts and figuresBull Emerg Trauma201423101102
  • FooteKDSeignourelPFernandezHHDual electrode thalamic deep brain stimulation for the treatment of posttraumatic and multiple sclerosis tremorNeurosurgery2006584 Suppl 2 ONS-280-5; discussion ONS-285-6
  • de HemptinneCRyapolova-WebbESAirELExaggerated phase-amplitude coupling in the primary motor cortex in Parkinson diseaseProc Natl Acad Sci U S A2013110124780478523471992
  • IssarNMHederaPPhibbsFTKonradPENeimatJSTreating post-traumatic tremor with deep brain stimulation: report of five casesParkinsonism Relat Disord201319121100110523953511
  • HasslerROreGDBricoloADieckmannGDolceGEEG and clinical arousal induced by bilateral long-term stimulation of pallidal systems in traumatic vigil comaElectroencephalogr Clin Neurophysiol19692776896904187363
  • SellalFHirschEBarthPBlondSMarescauxCA case of symptomatic hemidystonia improved by ventroposterolateral thalamic electrostimulationMov Disord1993845155188232364
  • SchiffNDGiacinoJTKalmarKBehavioural improvements with thalamic stimulation after severe traumatic brain injuryNature200744860060317671503
  • TsubokawaTYamamotoTKatayamaYHirayamaTMaejimaSMoriyaTDeep-brain stimulation in a persistent vegetative state: follow-up results and criteria for selection of candidatesBrain Inj199043153272252964
  • YamamotoTKatayamaYDeep brain stimulation therapy for the vegetative stateNeuropsychol Rehabil20051540641316350981
  • YamamotoTKatayamaYOshimaHFukayaCKawamataTTsubokawaTDeep brain stimulation therapy for a persistent vegetative stateActa Neurochir Suppl200279798211974994
  • YamamotoTKatayamaYKobayashiKKasaiMOshimaHFukayaCDBS therapy for persistent vegetative state: ten years follow-up resultsActa Neurochir Suppl200387151814518516
  • CapelleHHGripsEWeigelRPosttraumatic peripherally-induced dystonia and multifocal deep brain stimulation: case reportNeurosurgery2006593E702 discussion E70216955024
  • SonBCLeeSWChoiESSungJHHongJTMotor cortex stimulation for central pain following a traumatic brain injuryPain20061231–221021616616421
  • KuhnJLenartzDMaiJKHuffWKlosterkoetterJSturmVDisappearance of self-aggressive behavior in a brain-injured patient after deep brain stimulation of the hypothalamus: technical case reportNeurosurgery2008625E1182 discussion E118218580794
  • YamamotoTKatayamaYKobayashiKOshimaHFukayaCTsubokawaTDeep brain stimulation for the treatment of vegetative stateEur J Neurosci20103271145115121039954
  • ReeseRHerzogJFalkDSuccessful deep brain stimulation in a case of posttraumatic tremor and hemiparkinsonismMov Disord201126101954195521538523
  • YamamotoTKatayamaYObuchiTKobayashiKOshimaHFukayaCDeep brain stimulation and spinal cord stimulation for vegetative state and minimally conscious stateWorld Neurosurg2013803–4S30.e1S30.e922543046
  • FollettMATorres-RussottoDFollettKABilateral deep brain stimulation of the ventral intermediate nucleus of the thalamus for posttraumatic midbrain tremorNeuromodulation201417328929124024708
  • GiacinoJFinsJJMachadoASchiffNDCentral thalamic deep brain stimulation to promote recovery from chronic posttraumatic minimally conscious state: challenges and opportunitiesNeuromodulation201215433934922624587
  • ZhouLLinJKuiGZhangJYuYNeuroprotective effects of vagus nerve stimulation on traumatic brain injuryNeural Regen Res20149171585159125368644
  • CrownoverJWarmerAKRehabilomics concepts: an overview of genetic, proteomic and hormonal biomarkers in TBI recoveryWangKKWZhangZKobeissyFHBiomarkers of Brain Injury and Neurological Disorders1st edBoca RatonCRC Press2014236273
  • SiebnerHRBergmannTOBestmannSConsensus paper: combining transcranial stimulation with neuroimagingBrain Stimul200922588020633405
  • PéranPCataniSFaletta CaravassoCNoemiFSabatiniUFormisanoRSupplementary motor area activation is impaired in severe traumatic brain injury parkinsonismJ Neurotrauma201431764264824168468
  • PovlishockJTKatzDIUpdate of neuropathology and neurological recovery after traumatic brain injuryJ Head Trauma Rehabil2005201769415668572
  • FriedmanSDBrooksWMJungREQuantitative proton MRS predicts outcome after traumatic brain injuryNeurology 1995271384139110227622
  • BroggiGBrockSFranziniAGeminianiGA case of posttraumatic tremor treated by chronic stimulation of the thalamusMov Disord199382062088474491
  • LozanoAMMaybergHSGiacobbePHamaniCCraddockRCKennedySHSubcallosal cingulate gyrus deep brain stimulation for treatment-resistant depressionBiol Psychiatry20086446146718639234
  • MaybergHSLozanoAMVoonVDeep brain stimulation for treatment-resistant depressionNeuron20054565166015748841
  • OliveiraLFregniFPharmacological and electrical stimulation in chronic disorders of consciousness: new insights and future directionsBrain Inj201125431532721314279
  • KimiskidisVKValentinAKälviäinenRTranscranial magnetic stimulation for the diagnosis and treatment of epilepsyCurr Opin Neurol201427223624124553462
  • BauerPRKalitzinSZijlmansMSanderJWVisserGHCortical excitability as a potential clinical marker of epilepsy: a review of the clinical application of transcranial magnetic stimulationInt J Neural Syst2014242143000124475894