Next Article in Journal
7T MRI for Intracranial Vessel Wall Lesions and Its Associated Neurological Disorders: A Systematic Review
Previous Article in Journal
A High Methylation Level of a Novel −284 bp CpG Island in the RAMP1 Gene Promoter Is Potentially Associated with Migraine in Women
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Current Clinical Trials in Traumatic Brain Injury

1
Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
2
Centre for Trauma Sciences Research, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
3
Surgical Reconstruction and Microbiology Research Centre, National Institute for Health Research, Queen Elizabeth Hospital, Birmingham B15 2TH, UK
Brain Sci. 2022, 12(5), 527; https://doi.org/10.3390/brainsci12050527
Submission received: 7 March 2022 / Revised: 13 April 2022 / Accepted: 19 April 2022 / Published: 21 April 2022
(This article belongs to the Section Molecular and Cellular Neuroscience)

Abstract

:
Traumatic brain injury (TBI) is one of the leading causes of morbidity, disability and mortality across all age groups globally. Currently, only palliative treatments exist, but these are suboptimal and do little to combat the progressive damage to the brain that occurs after a TBI. However, multiple experimental treatments are currently available that target the primary and secondary biochemical and cellular changes that occur after a TBI. Some of these drugs have progressed to clinical trials and are currently being evaluated for their therapeutic benefits in TBI patients. The aim of this study was to identify which drugs are currently being evaluated in clinical trials for TBI. A search of ClinicalTrials.gov was performed on 3 December 2021 and all clinical trials that mentioned “TBI” OR “traumatic brain injury” AND “drug” were searched, revealing 362 registered trials. Of the trials, 46 were excluded due to the drug not being mentioned, leaving 138 that were completed and 116 that were withdrawn. Although the studies included 267,298 TBI patients, the average number of patients per study was 865 with a range of 5–200,000. Of the completed studies, 125 different drugs were tested in TBI patients but only 7 drugs were used in more than three studies, including amantadine, botulinum toxin A and tranexamic acid (TXA). However, previous clinical studies using these seven drugs showed variable results. The current study concludes that clinical trials in TBI have to be carefully conducted so as to reduce variability across studies, since the severity of TBI and timing of therapeutic interventions were key aspects of trial success.

1. Introduction

Traumatic brain injury (TBI) is one of the leading causes of morbidity, disability and mortality across all age groups [1,2]. The biggest causes of TBI are motor vehicle accidents, falls and interpersonal violence. More than 50 million individuals suffer a TBI each year and there are approximately 3.2 million TBI survivors who experience post-TBI complications, which include neurological and psychological problems as well as long-term disability [3,4,5]. In the UK alone, clinical management of TBI costs the economy GBP 15 billion per year, representing a significant burden on the UK economy.
TBI is classified into different categories including closed head, penetrating and explosive blast TBI. TBI patients suffer a variety of symptoms including headache, nausea, seizures, amnesia, aggression and anxiety. These symptoms can appear within seconds after a TBI, and some of the effects can last months to years [1,6]. Closed head TBI typically presents after blunt trauma impact mainly from motor vehicle accidents, falls and sports. Penetrating TBI occurs when a foreign body penetrates the skull and traverses through into the brain parenchyma. Explosive blast TBI occurs primarily in war-related TBI, where the rapid shock waves of a blast transmit kinetic energy from the skull and into the parenchyma causing deformation of the brain. All TBIs result in damage to the brain tissues, including the vasculature, diffuse axonal injury, compromised blood brain barrier and cerebral oedema.
Damage to the brain occurs both from the primary and secondary injury processes. The primary injury results from the direct mechanical forces acting on the brain during the initial insult whilst the secondary injury results from tissue and cell damages that follow the initial insult. The primary injury causes both focal and diffuse consequences which leads to tissue necrosis, hematomas and intracerebral hemorrhages, and eventually damages axons, oligodendrocytes and the vasculature, leading to oedema and ischemic brain damage [7,8,9,10]. The secondary brain injury occurs as a result of the biochemical, cellular and physiological changes that follow a TBI. For example, excitotoxicity, mitochondrial dysfunction, oxidative stress, lipid peroxidation, neuroinflammation, axon degeneration and apoptosis all occur after a TBI and contribute to the secondary injury processes [11].
Many of the treatment regimens for TBI focus on stabilization of the injured brain and prevention of secondary injury processes. Since secondary injury characteristics develop progressively, a therapeutic window of opportunity exists in order to protect further loss of neurons and glia as well as targeting excitotoxicity, inflammation, oxidative stress and apoptosis. An increased understanding of the clinical characteristics and the underlying complex pathophysiological mechanisms of TBI has led to the development of several novel and promising therapeutic approaches that have shown positive effects in preclinical studies. Many of these drugs have progressed from preclinical studies into clinical evaluation of their usefulness in treating TBI.
Some of the long-term consequences of TBI include problems with balance, motricity, headaches, cognitive and behavioral problems and post-traumatic epilepsy [12,13,14,15]. Many patients also suffer from anxiety or depression, and management of these symptoms are top priorities among the most frequently perceived needs [16,17,18]. About a third of TBI subjects were lower moderate to severely disabled, requiring help with daily living activities, whilst a third were at an upper moderate disability level, meaning that they were independent inside and outside their homes but had reduced work capacity, social, leisure and family or friendship disruption [16,19,20]. Counteracting these long-term consequences of TBI must be factored into future TBI clinical trials.
The aim of this review is to present an overview of the clinical trials that were registered with ClinicalTrials.gov on 3 December 2021 and present data on the number of trials that have been completed and the number of patients in each trial, as well as reviewing the drug treatments that are being tested. The review also aims to provide some guidelines on improving future clinical trial design, which need to be considered in this area of unmet medical need.

2. Trials Registered in ClinicalTrials

The ClinicalTrials.gov (accessed on 3 December 2021) database was searched with the terms “TBI” OR “traumatic brain injury” AND “drug”. As of 3 December 2021, there were 361 trials registered on Clinical Trials.gov that were returned after searching for “TBI” OR “traumatic brain injury” AND “drug” (Figure 1). Of all studies retrieved, 46 were excluded or not relevant due to the drug not being mentioned. Of the trials listed, 138 were completed, 116 were withdrawn, suspended or the status was unknown, 46 were still recruiting, 5 were active but not yet recruiting, 13 were not recruiting and 2 were enrolling by invitation (Figure 1).
Although all of the studies included 267,298 patients, the average number of patients was 865 per study with a range of 5–200,000 and a median of 50 patients/study.

2.1. Completed Studies

Of the studies completed, the highest numbers were completed between 2016 (16) and 2017 (17), with fewer studies completed since then (Figure 2). The total number of patients in these studies was 40,947, with an average of 1742 patients/study, a range of 10–14,959 and a median of 130 patients. Patients were stratified into 46 different age groups in the 138 completed studies, ranging from categories such as child, adult, older adult and anywhere between 2 to 100 years old (Figure 3). The largest age groups for enrollment into a TBI trial were 18 years and older (3600 participants), 18 to 60 years old (13,043 participants) and 18 to 65 (14,959 participants) (Figure 3).
Of the completed studies, 24 were early Phase I, 8 were Phase I/II, 35 were Phase II, 5 were Phase II/III, 20 were Phase III, 22 were Phase IV and 24 were not applicable to be assigned to a phase of study (Figure 4).
Between the completed studies, 125 different drugs for TBI were evaluated. However, only seven drugs were used in three or more studies across the different clinical phases of study, including amantadine, botulinum toxin type A, hyperbaric oxygen, methylphenidate, NNZ-2566, Rivastigmine and tranexamic acid (TXA) (Figure 5).

2.2. Phase IV Studies

There are currently 18 drugs in Phase IV clinical trials. These are drugs that have been approved previously but are being investigated in studies where the side effects caused over time are being evaluated. These include Dipeptiven, propranolol, methylphenidate, rHGF, Lisdexamfetamine, Venlafaxine, Rivastigmine, citalopram (celexa), Doxycycline, simvastatin, Enoxaparin, Levetiracetam, Androgel (testosterone), amantadine, botulinum toxin type A, intravenous acetaminophen, Nuedexta and 20% mannitol.

3. Characteristics of Completed Studies

3.1. Completed Studies with Results

There were only 44 clinical trials in TBI that have been completed and where results are available. Table 1 provides a summary of these studies, their primary outcomes and the results posted on ClinicalTrials.gov (accessed on 3 December 2021). Surprisingly, only six study results have been published (NCT00313716, NCT01322048, NCT02012582, NCT01750268, NCT02270736, NCT01463033). Of the 44 studies, 30 showed no statistical difference between treatment and controls, 7 studies showed better results in the treatment arm but lacked statistical data, 4 studies had only a single arm and so comparisons could not be made, 2 studies showed worse outcomes in the treatment arm, whilst only 1 study demonstrated significantly better results in the treatment arm (NCT02270736). Twenty-four studies failed to meet their recruitment targets whilst seventeen studies recruited more patients than posted on ClinicalTrials.gov (accessed on 3 December 2021).

3.2. Summary of Evidence for Drugs Appearing in Three or More TBI Studies

3.2.1. Amantadine

Amantadine is a dopaminergic agent and is an antagonist of N-methyl-D-aspartate (NMDA), approved by the FDA for use in the prevention of influenza and Parkinson’s disease [21]. Amantadine is most commonly prescribed for patients with disorders of consciousness and who are undergoing inpatient neurorehabilitation, although its mechanism of action is unclear. However, several clinical trials have shown positive effects with amantadine in terms of neurobehavioral recovery, cognitive function and improved disability rating scores [22,23,24]. Other studies have either shown no benefits of amantadine or some positive benefits; however, these studies only had 20–25 participants [25,26]. In a study in 2018, the use of amantadine to improve cognitive performance was not supported in a multi-site, randomized controlled trial (RCT) [27]. Although amantadine is well tolerated, definite improvements in cognition and neurobehavioral recovery after TBI remains to be reported.

3.2.2. Botulinum Toxin Type A

Botulinum toxin type A (BoNT-A) is a potent neurotoxin produced by the Gram negative aerobic bacterium Clostridium botulinum. BoNT-A has been used as a pharmacological treatment for the management of spasticity and exerts its effects by binding pre-synaptically to high-affinity recognition sites on the cholinergic nerve terminals. This inhibits the release of acetylcholine, causing temporary neuromuscular blockade and muscle relaxation [28,29]. The effects of BoNT-A are temporary, and neurotransmission slowly resumes as the neuromuscular junction recovers with time [28]. BoNT-A has been used to treat upper limb spasticity after stroke or TBI [29,30,31,32,33]. Clinical studies have shown that a significant reduction in muscle tone is observed as early as one week, with peak effects at 4–6 weeks after injection, waning thereafter [34,35,36].

3.2.3. Hyperbaric Oxygen

Hyperbaric oxygen therapy (HBOT) requires the inhalation of 100% oxygen under a pressure that is greater than 1 atmosphere. Experimental studies of HBOT after TBI demonstrates inhibition of apoptosis, suppression of inflammation, protection of the blood–brain barrier, and promotion of angiogenesis and neurogenesis, with a range of HBOT treatments from 1.5 atmospheres to 3 atmospheres and up to 90 min twice daily for 40 days to 45 min for two sessions [37]. HBOT in humans after brain injury can correct brain anoxia and craniocerebral oedema, reduce intracranial pressure and improve neurological function, prognosis and quality of life in patients [38,39]. In RCTs, HBOT for severe TBI demonstrated higher GCS scores and lower National Institutes of Health Stroke Scale (NIHSS) [40]. The study concluded that higher GCS on admission, tracheotomy status and first HBOT duration were independent prognostic factors in patients with severe TBI [40]. HBOT treatment also led to improved cognitive function in TBI patients [41], however these studies post-TBI are limited and hence further RCTs are required.

3.2.4. Methylphenidate

TBI results in alterations in the chemistry and structure of brain cells and long-term changes in the levels of neurotransmitters. Reduced serotonin and catecholamine are related to TBI-associated neurological comorbidities [42]. Methylphenidate, a psychostimulant which blocks the reuptake of norepinephrine and dopamine into the presynaptic neuron, is used to treat narcolepsy and attention deficit hyperactivity disorder (ADHD) in children [43] but is potentially beneficial against TBI-associated neurological sequelae [44]. Although the exact mechanism is not clear, it is thought to activate the brainstem arousal system, the cortex and subcortical regions such as the thalamus, and produce its stimulant effect on the brain. In a study of ten RCTs, methylphenidate showed significant improvements in enhancing vigilance-associated attention (i.e., selective, sustained and divided attention) in TBI patients, but no significant effects on memory or processing speed were noted [45]. However, most of these previous studies included TBIs with a wide range of severities, age, small samples sizes or use of an open-label design and hence further adequately powered, well-designed, double-blind, placebo-controlled RCTs are required to reach definitive conclusions regarding the use of methylphenidate in TBI [44,46,47,48,49,50,51,52].

3.2.5. NNZ-2566

NNZ-2566 is an analogue of endogenous tripeptide glycine-proline-glutamate with improved stability, and after penetrating ballistic-like brain injury has been shown to be neuroprotective as well as improved motor function and reduced incidence, frequency and duration of post-injury seizures [53,54,55,56]. NZZ-256 exerts anti-inflammatory properties, as it reduces injury-induced increases in pro-inflammatory cytokines, suggesting that NZZ-256 exerts its influence through modulation of the immune response after TBI [57]. Despite the limited number of studies with NNZ-2566 in TBI, Neuren Pharmaceuticals has conducted Phase I and II clinical trials in patients with TBI.

3.2.6. Rivastigmine

Rivastigmine treatment after severe closed head injury reduced cerebral oedema and accelerated motor and cognitive function recovery, effects that were mediated by increased cholinergic activity at both muscarinic and nicotinic receptors [58]. There were some initial Phase I studies of Rivastigmine in TBI showing that it was safe [59,60]; however, an RCT study showed that 17 patients had to withdraw due to adverse events, with 69 patients completing the trial [60]. Clinical interviews, however, failed to show statistically significant positive benefits of Rivastigmine in vigilance tests in the 69 patients that completed the trial [60]. Moreover, a recent Phase III study failed to show any benefits on cognitive function using a Rivastigmine patch in veterans with TBI [61].

3.2.7. Tranexamic Acid (TXA)

Uncontrolled hemorrhage after trauma is a cause of early mortality in major trauma, accounting for 30–40% of all deaths. TBI is associated with intracranial bleeds in 25 to 40%, 3 to 12%, and 0.2% of severe, moderate and mild TBI patients, respectively [62]. In addition, one-third of TBI patients suffer from coagulopathy, requiring treatment with antifibrinolytic agents such as TXA [63,64,65]. TXA can therefore reduce bleeding and mortality without adverse effects. TXA is a lysine analogue with anti-fibrinolytic actions and competitively binds to lysine sites of plasminogen and plasmin, inhibiting the binding of plasmin to fibrin and subsequently preventing fibrinolysis [66,67].
Despite numerous studies showing some benefits of TXA in TBI, a systematic review of nine RCTs with 14,747 patients found no statistical benefits on mortality or disability after TBI [68]. This was also supported by an Editorial from the Journal “Intensive Care Medicine”, pointing out that the systematic review by Lawati (2021) [68] analyzed all causes of mortality, whereas the CRASH-3 study used “head injury-related death” as a definition to analyze mortality, and this could be prone to information bias [69]. Another study found that prehospital administration of TXA in all TBI patients significantly increased the risk of 30-day mortality rates, with a higher risk in those with severe isolated TBI [70]. Given the current evidence on TXA, further studies are required to clear up whether TXA is beneficial in TBI patients or not.

3.3. Ongoing Studies

Several TBI studies are currently ongoing, with 3 studies that are active but not recruiting (Table 2), 37 studies that are currently recruiting (Table 3) and 13 studies are yet to recruit (Table 4). The biggest study planned is the Phase III CRASH-4 trial, with 10,000 participants to be enrolled onto the study, a continuation of the CRASH trials to evaluate the effect of the antifibrinolytic agent, tranexamic acid, in mild head injury in older adults (50 years and older). Other studies of note are NCT04588311, which is a Phase III study that will evaluate the effect of erythropoietin-alpha in preventing mortality and reducing severe disability not only in TBI patients but also other severe trauma patients. NCT03061565 will compare the effects of erythropoietin in reducing mortality over the longer term and hopes to recruit over 600 patients.
Other drugs currently being tested in Phase III studies include Biperiden Lactate to reduce post-TBI epilepsy, dexamethasone to reduce pericontusional oedema, Dalteparin to prevent venous thromboembolism, NT201 to reduce lower limb spasticity after TBI, inhaled nitric oxide to reduce secondary brain damage, Nucleo CMP Forte to protect against glutamate toxicity in children, and the effect of citoflavin to improve cerebral blood flow, restore impaired consciousness and improve cognitive outcomes (Table 3).

3.4. Summary of Drugs to Be Used in Large TBI Studies That Are Currently Active

Examples of some of the largest studies (>500 participants) that are currently registered active on ClinicalTrials.gov (accessed on 3 December 2021) include TXA, erythropoietin, phenytoin sodium, Dalteparin, propranolol, NT201 and Dexamethasone. Section 3.2.7. contains a summary of the results thus far with tranexamic acid in TBI and hence they are not discussed here.

3.4.1. Erythropoietin

Erythropoietin (EPO) is a hemopoietic growth factor with neurocytoprotective effects and is normally produced in the spleen, liver, bone marrow, lung and brain [71]. Although EPO is mainly used in conditions where there is impaired red blood cell production, it is neuroprotective and neuroregenerative by reducing apoptosis, inflammation, oxidative stress and excitotoxicity [72,73]. EPO reduces lesion volume and improves neurobehavioral outcomes after TBI [74]. The clinical benefits of EPO in TBI were later realized but data were conflicting [75,76]. One systematic review suggested that EPO reduced overall mortality and shortened hospitalization time but did not improve neurological outcomes [77]. A more recent systematic review involving seven randomized controlled trials (RCTs) and 1197 TBI patients (611 treated with EPO) found no improvements in acute hospital or short-term mortality but did show a significant improvement in mid-term (6 month) follow-up survival rates [78]. Disappointingly, the study found that EPO was not associated with neurological functional improvements [78].
What is also clear is that there were different EPO treatment doses used across the seven studies, which may account for variability. For example, EPO administration ranged from 500 IU/kg to 40,000 IU, subcutaneously or intravenously injected, some with repeated doses at days and weeks after the initial doses [78]. The majority of the studies administered EPO within 6 h although one study reported up to 24 h. The patient populations were also varied, with some that were reported as severe and some that were moderate in severity. All of these differences could account for the variability in the reported outcomes, and hence uniformity will be important in future studies.

3.4.2. Phenytoin Sodium

Phenytoin is a widely used anti-epileptic drug used to control post-traumatic seizure prophylaxis. The use of anti-epileptic drugs in TBI remains a point of contention. However, it is recognized that post-TBI seizures develop in 12% of severe TBI cases and that phenytoin treatment reduces this to 3.6% [79]. Current guidelines for post-TBI seizures focus on control efficacy, for which phenytoin and Levetiracetam is commonly used. However, phenytoin has several complications which limits its use, including imbalance and dizziness [80]. In a recent systematic review, the authors failed to identify differences between phenytoin and Levetiracetam in any of the outcomes after TBI including early seizures, stating that further well-powered RCTs are required to reach definitive conclusions on the benefits of phenytoin after TBI [81]. In this regard, the MAST trial (NCT04573803) plans to recruit 1649 participants in a Phase III trial to assess phenytoin and Levetiracetam, and aims to answer whether a shorter or a longer course of anti-epileptic drugs prevent further seizures in patients that have started having seizures after TBI, as well as whether a 7-day course of phenytoin and Levetiracetam should be used in patients with serious TBI to prevent seizures from starting. Results from this trial are eagerly awaited and will go some way to answer whether anti-epileptic drugs should be used in TBI patients.

3.4.3. Dalteparin

TBI patients are at high risk of venous thromboembolic events (VTE), defined as either deep vein thrombosis (DVT) or pulmonary embolism (PE). The risk of baseline VTE is approximately 5%, and this increases to 30–60% in patients with TBI [82,83]. Commonly, low molecular weight heparin or unfractionated heparin is used to prevent VTE complications; however, heparin can increase the risk of expansion of intracranial hemorrhages with VTE prophylaxis [82]. In an early study that retrospectively compared VTE rates between Dalteparin and Enoxaparin, no significant differences between the two drugs was observed. The groups treated with Dalteparin tended to be more severe TBI patients and the study concluded that VTE prophylaxis in TBI patients offered high levels of protection against VTE with an extremely low risk of expansion of intracranial hemorrhages [84].
In a recent systematic review that included 21 studies, VTE prophylaxis did not lead to TBI progression and VTE prophylaxis with 24–72 h after TBI was safe in patients with stable injuries [85]. There was also no relationship between hemorrhagic progression and timing of VEP prophylaxis [85]. In an international comparative study in the Netherlands and the USA, almost 80% of trauma patients received VTE prophylaxis, with a greater proportion of patients with VTE afflicted by TBI [86]. VTE occurred in 75–81% of patients despite receiving adequate VTE prophylaxis and within 48 h of injury. This may suggest that patients developing a VTE are at such a high risk that even with chemical prophylaxis treatment, it is not sufficient or is unable to be started early enough post injury to demonstrate a beneficial effect. Future clinical trials will need to address these concerns.

3.4.4. Propanolol

Severe TBI causes a surge in catecholamines such as epinephrine and norepinephrine, and these remain elevated in patients with persistent coma or who are moribund [87]. In those with TBI, plasma norepinephrine levels at 48 hours post injury are predictive of Glasgow Coma Scale (GCS) at 1 week, survival, the number of ventilator days and the length of stay (LOS) [88]. Systemically, this sympathetic surge causes tachycardia, tachypnea, hypertension and hyperpyrexia with associated motor features such as agitation and dystonia [89]. TBI severity also correlates with decreased heart rate, and persistent sympathetic hyperactivity is also associated with increased length of stay in intensive care units, lower cognitive abilities and higher cognitive fatigue [90,91,92].
The use of β-blockade, such as the non-selective β-blocker propranolol in pre-clinical mouse studies, reduced brain oedema, improved neurological outcomes, increased cerebral perfusion and decreased cerebral hypoxia [93,94,95]. Propranolol also reduced the maximum intensity of agitated episodes as well as reduced aggressive behavior months after TBI [96,97]. This was followed up by two parallel clinical trials (NCT01202110, NCT01343329) which used early treatment with propranolol in TBI and reported improvements in short-term endpoints such as heart rate [98]. In further retrospective studies, β-blockade after TBI conveyed 4–23% improved mortality rates [99]. The DASH after TBI trial (NCT01322048), a randomized, double-blind placebo-controlled trial with 48 patients in total (21 with TBI and 26 with placebo), posted some results in 2017 which showed that the study did not detect significant changes in primary outcomes of ventilator-free days. Therefore, further high-quality studies are required to evaluate the potential benefits of propranolol in TBI patients.

3.4.5. NT201

NT201 is botulinum toxin type A (BoNT-A) and was mentioned earlier in this review. Initial studies in patients with brain injury or cerebral palsy offered significant reductions in spasticity in elbow, wrist, fingers and ankle muscles receiving high doses of NT201 [30,31,32,33,34]. The PATTERN study (NCT03992404) is currently still recruiting patients in a study to compare the efficacy and safety of NT201 in the treatment of lower limb spasticity caused by stroke or TBI, with a planned recruitment of 600 participants in a randomized parallel double-blind study. The primary outcomes for this trial will be the modified Ashworth scale–Bohannon (MAS) ankle score at 4–6 weeks with a co-primary outcome of global impression of change scale (GICS) at 4–6 weeks.

3.4.6. Dexamethasone

Cerebral oedema after TBI is a serious complication which corticosteroids, including glucocorticoids, can ameliorate effectively [100]. Dexamethasone is a corticosteroid that acts on the glucocorticoid receptor and efficiently reduces blood–brain barrier (BBB) permeability. However, dexamethasone does not readily cross the BBB and the mechanisms by which dexamethasone is neuroprotective are still not understood. The CRASH study in 2004, which was a multi-center trial to assess the use of corticosteroids acutely after head injury, concluded that the use of steroids in the acute period after injury caused more harm than good and are not recommended in head injury [101]. However, dexamethasone is commonly administered to patients undergoing a variety of neurosurgical procedures [100]. In TBI patients, dexamethasone treatment significantly reduced the volume of vasogenic oedema, decreased the apparent diffusion coefficient and increased fractional anisotropy, suggesting beneficial effects of this drug in TBI patients [102]. However, the study was a prospective observational study with only 30 TBI patients, and so larger studies are required to confirm these results.
The DEXCON-TBI study (NCT04303065) is a multicenter, randomized triple-blind placebo-controlled study that will quantify the effects of the administration of dexamethasone on the prognosis of TBI patients with brain contusions and pericontusional oedema. The primary outcome for the trial is improvements in the Glasgow scale outcome extended (GOSE) measure with a number of secondary outcomes. The study will recruit 600 participants with a short and descending course of dexamethasone. The results of this study are eagerly awaited and will determine if dexamethasone is likely to confer benefits when administered acutely.

4. Discussion

This study reports that there were a total of 361 registered clinical trials listed on ClinicalTrials.gov (accessed on 3 December 2021) containing the search terms “traumatic brain injury” OR “TBI” AND “drug” as of 3 December 2021. Of the trials listed, 138 were completed, 116 were withdrawn, suspended or the status was unknown, 46 were still recruiting, 5 were active but not yet recruiting, 13 were not recruiting and 2 were enrolling by invitation. The average number of patients recruited per study was 865 (range 5–200,000) with a median of 50 patients/study. Of the completed studies, 125 different drugs were reported to be evaluated. Only seven drugs appeared in three or more trials, which represented the most promising treatment options for TBI and included amantadine, botulinum toxin type A, hyperbaric oxygen, methylphenidate, NNZ-2566, Rivastigmine and TXA.
Only 44 of the 138 completed trials posted results on ClinicalTrials.gov (accessed on 3 December 2021) and of these, only 6 studies have been published in a peer reviewed journal. Interestingly, the published studies were only studies that were positive or contained some aspect of positive data that could be reported. All of the other studies with results were largely negative and remained unpublished in peer reviewed journals. Of the studies that posted results, 30 showed no statistical difference between the treatment arm and the placebo/control arm, 2 studies showed worse outcomes and only 1 study demonstrated statistically significant results. In addition, seven of the studies showed better results in the treatment arm but lacked statistical analysis, whilst four studies only included a single arm, presumably due to low study recruitment, and so comparisons were not possible. A significant number of studies also failed to meet their recruitment targets and so a number of studies were underpowered. Surprisingly, some studies recruited more patients than was posted on ClinicalTrials.gov (accessed on 3 December 2021).
Although TXA appears to be the most likely to translate to the clinic, as alluded to in Section 3.2.7 of this review, there is some debate about the benefits of TXA in TBI with a recent systematic review of nine RCTs showing no benefits in mortality or disability [68]. The CRASH-3 study itself enrolled 9,202 head injury patients within 3 h of injury with a GCS of <12 or any intracranial bleeding on computed tomography. The study found no statistical difference in the primary outcome of head injury related death (18.5% with TXA and 19.8% with placebo) (relative risk (RR): 0.94, 95% CI 0.86–1.02). However, there were significant differences in subgroups that were less severely injured (i.e., when those with GCS = 3 or bilateral fixed pupils were excluded (RR: 0.89, 95% CI: 0.80 to 1.00) and in the GCS 9–15 subgroup (RR: 0.78, 95% CI: 0.64 to 0.95), or those that were treated earlier (p = 0.005 for time effect) [103]. In these less severely injured patients, it would be expected that they would have the highest mortality benefit and so it may be reasonable to give TXA to this subgroup; however, there is not enough evidence to consider this as standard practice. Furthermore, in the subgroup analysis, there was no indication of the all-cause mortality rate. In contrast, TXA also increased death from all other causes, although the result was not statistically significant (RR: 1.31; 95% CI 0.93–1.85). Moreover, the CRASH-3 trial reported that disability remained unchanged, and so based on the evidence, the trial can be classed as negative, since the primary outcome was not met, and neither was disability affected in favor of TXA. Although it has been claimed that the study is underpowered despite involving over 9000 participants, an official video claiming that TXA “could save 10 s of thousands of lives” are unfounded [104].
Of the studies that were completed and published in peer-reviewed journals, there were other studies that failed to meet the primary outcomes but reported statistically significant data in other outcomes or subgroups. Whether this is helpful in improving the design of future clinical trials remains to be seen. However, these studies highlight several aspects of future clinical trial design that need to be taken into account. Although a variety of drugs are being analyzed in clinical trials in TBI, the variability in the reported study results warrants some discussion and refinement in the design of future clinical trials. One major issue within clinical trials of TBI is that all TBI patients, including severe, moderate and mild TBI, are often included. There are clearly significant pathological differences between severe, moderate and mild TBI and therapeutics will have different levels of benefits depending on injury severity. Even using the GCS to stratify TBI patients, heterogeneity is inevitable, since multiple causes may contribute to the same GCS score including diffuse axonal injury, diffuse swelling, contusion and hematoma.
TBIs are also heterogenous (e.g., extradural/subdural, diffuse axons or focal, etc.), and hence the outcome of potential treatments are governed by multiple factors including injury location, physiology and whether the TBI is associated with extracranial injuries. Approximately a quarter of “mild” head injury patients do not return to work and >80% of patients have associated problems even after one year post-TBI, calling into question the term “mild TBI”. Therefore, future studies should stratify patients carefully prior to enrollment in a clinical trial and target only the same patient severities.
Another key consideration in the design of clinical trials is timing of therapeutic intervention. Some treatments will need to be given as early as possible whilst other treatments can be given later. For example, treatments for oedema and to control bleeding are likely to be required immediately after injury and probably best given during the prehospital period to control these adverse events. Neuroprotective treatments may also need to be given within minutes of the injury, which is difficult, since neurons begin to die within minutes of TBI and delays may mask a real neuroprotective effect of a given drug.
Most acute TBI studies are conducted in intensive care which is a safe and controlled environment. However, secondary injuries are likely to occur during the prehospital period where hypoxia, hypotension and expanding hematoma may cause the greatest amounts of neurological damage and where therapeutic interventions may have the best impact. Hence, future clinicals trials should consider studies in the prehospital environment.
One significant issue is that not all clinical trials are published as manuscripts. Only 6 studies were published from amongst the 44 clinical trials that had results posted on ClinicalTrials.gov (accessed on 3 December 2021). Interestingly, these were either studies that were positive or had some positive data that could be reported. It is therefore a concern that only studies with positive data are being published. All of these clinical trials should be written up and published in peer reviewed journals whether positive or negative. This is especially true since significant amounts of resources, manpower and time have been spent on such clinical trials. The studies themselves have value in the sense that it informs other researchers of particular treatments or the design of better studies to obtain unequivocal data regarding the efficacy of a particular compound. The issue of positive publication bias has been highlighted by many and may result in bias in meta-analyses, leading to distortion of literature and misleading researchers, doctors and even policymakers in their decision making [103,104,105,106]. Therefore, all data, be it positive or negative, should be published as long as the study has been performed rigorously and adheres to high-quality standards in study design and analysis. Not publishing the data may be deemed unethical.

Future Clinical Trial Designs

New clinical trial designs are being recommended in TBI to enhance therapy development [107]. These include comparative effectiveness studies such as those under way in adults and children, CENTER-TBI and ADAPT studies, respectively [108,109]; adaptive trial design where computer algorithms are used to randomize patients in a blinded fashion, those that show favorable effects, rather than randomizing to the test and placebo/control groups equally [110]; continuous data acquisition using electronic medical records rather than data entry of hourly physiological findings [111,112]; and big-data approaches to identify associations and treatment efficacies [113]. All of these recommendations need to be considered in future clinical trial designs to improve the translation of new therapies into the clinic for the benefit of TBI patients.

Funding

This research received no external funding.

Institutional Review Board Statement

Ethical review and approval were waived for this study as it is a review of published literature.

Informed Consent Statement

Not applicable since patients or members of the public were not involved in the design, conduct of this study, or reporting of this research.

Data Availability Statement

All data generated as part of this study are included in the article.

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Bruns, J., Jr.; Hauser, W.A. The epidemiology of traumatic brain injury: A review. Epilepsia 2003, 44, 2–10. [Google Scholar] [CrossRef] [PubMed]
  2. Dewan, M.C.; Rattani, A.; Gupta, S.; Baticulon, R.E.; Hung, Y.C.; Punchak, M.; Agrawal, A.; Adeleye, A.O.; Shrime, M.G.; Rubiano, A.M.; et al. Estimating the global incidence of traumatic brain injury. J. Neurosurg. 2018, 130, 1080–1097. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Maas, A.I.R.; Menon, D.K.; Adelson, P.D.; Andelic, N.; Bell, M.J.; Belli, A.; Bragge, P.; Brazinova, A.; Buki, A.; Chesnut, R.M.; et al. Traumatic brain injury: Integrated approaches to improve prevention, clinical care, and research. Lancet Neurol. 2017, 16, 987–1048. [Google Scholar] [CrossRef] [Green Version]
  4. Zaloshnja, E.; Miller, T.; Langlois, J.A.; Selassie, A.W. Prevalence of long-term disability from traumatic brain injury in the civilian population of the United States, 2005. J. Head Trauma Rehabil. 2008, 23, 394–400. [Google Scholar] [CrossRef]
  5. Bazarian, J.J.; Cernak, I.; Noble-Haeusslein, L.; Potolicchio, S.; Temkin, N. Long-term neurologic outcomes after traumatic brain injury. J. Head Trauma Rehabil. 2009, 24, 439–451. [Google Scholar] [CrossRef]
  6. Andriessen, T.M.; Jacobs, B.; Vos, P.E. Clinical characteristics and pathophysiological mechanisms of focal and diffuse traumatic brain injury. J. Cell. Mol. Med. 2010, 14, 2381–2392. [Google Scholar] [CrossRef] [Green Version]
  7. Skandsen, T.; Finnanger, T.G.; Andersson, S.; Lydersen, S.; Brunner, J.F.; Vik, A. Cognitive impairment 3 months after moderate and severe traumatic brain injury: A prospective follow-up study. Arch. Phys. Med. Rehabil. 2010, 91, 1904–1913. [Google Scholar] [CrossRef]
  8. Glaesser, J.; Neuner, F.; Lutgehetmann, R.; Schmidt, R.; Elbert, T. Posttraumatic Stress Disorder in patients with traumatic brain injury. BMC Psychiatry 2004, 4, 5. [Google Scholar] [CrossRef] [Green Version]
  9. Smith, D.H.; Meaney, D.F.; Shull, W.H. Diffuse axonal injury in head trauma. J. Head Trauma Rehabil. 2003, 18, 307–316. [Google Scholar] [CrossRef] [Green Version]
  10. Saatman, K.E.; Duhaime, A.C.; Bullock, R.; Maas, A.I.; Valadka, A.; Manley, G.T.; Workshop Scientific, T.; Advisory Panel, M. Classification of traumatic brain injury for targeted therapies. J. Neurotrauma 2008, 25, 719–738. [Google Scholar] [CrossRef] [Green Version]
  11. Ray, S.K.; Dixon, C.E.; Banik, N.L. Molecular mechanisms in the pathogenesis of traumatic brain injury. Histol. Histopathol. 2002, 17, 1137–1152. [Google Scholar] [CrossRef]
  12. Rosenfeld, J.V.; Maas, A.I.; Bragge, P.; Morganti-Kossmann, M.C.; Manley, G.T.; Gruen, R.L. Early management of severe traumatic brain injury. Lancet 2012, 380, 1088–1098. [Google Scholar] [CrossRef]
  13. Thomsen, I.V. Late outcome of very severe blunt head trauma: A 10–15 year second follow-up. J. Neurol. Neurosurg. Psychiatry 1984, 47, 260–268. [Google Scholar] [CrossRef] [Green Version]
  14. DeGrauw, X.; Thurman, D.; Xu, L.; Kancherla, V.; DeGrauw, T. Epidemiology of traumatic brain injury-associated epilepsy and early use of anti-epilepsy drugs: An analysis of insurance claims data, 2004–2014. Epilepsy Res. 2018, 146, 41–49. [Google Scholar] [CrossRef]
  15. Ferguson, P.L.; Pickelsimer, E.E.; Corrigan, J.D.; Bogner, J.A.; Wald, M. Prevalence of traumatic brain injury among prisoners in South Carolina. J. Head Trauma Rehabil. 2012, 27, E11–E20. [Google Scholar] [CrossRef]
  16. Draper, K.; Ponsford, J.; Schönberger, M. Psychosocial and emotional outcomes 10 years following traumatic brain injury. J. Head Trauma Rehabil. 2007, 22, 278–287. [Google Scholar] [CrossRef]
  17. Corrigan, J.D.; Whiteneck, G.; Mellick, D. Perceived needs following traumatic brain injury. J. Head Trauma Rehabil. 2004, 19, 205–216. [Google Scholar] [CrossRef]
  18. Andelic, N.; Soberg, H.L.; Berntsen, S.; Sigurdardottir, S.; Roe, C. Self-perceived health care needs and delivery of health care services 5 years after moderate-to-severe traumatic brain injury. PMR 2014, 6, 1013–1021. [Google Scholar] [CrossRef]
  19. Andelic, N.; Hammergren, N.; Bautz-Holter, E.; Sveen, U.; Brunborg, C.; Røe, C. Functional outcome and health-related quality of life 10 years after moderate-to-severe traumatic brain injury. Acta Neurol. Scand. 2009, 120, 16–23. [Google Scholar] [CrossRef]
  20. Andelic, N.; Howe, E.I.; Hellstrøm, T.; Sanchez, M.F.; Lu, J.; Løvstad, M.; Røe, C. Disability and quality of life 20 years after traumatic brain injury. Brain Behav. 2018, 8, e01018. [Google Scholar] [CrossRef]
  21. Warden, D.L.; Gordon, B.; McAllister, T.W.; Silver, J.M.; Barth, J.T.; Bruns, J.; Drake, A.; Gentry, T.; Jagoda, A.; Katz, D.I.; et al. Guidelines for the pharmacologic treatment of neurobehavioral sequelae of traumatic brain injury. J. Neurotrauma 2006, 23, 1468–1501. [Google Scholar] [CrossRef] [Green Version]
  22. Meythaler, J.M.; Brunner, R.C.; Johnson, A.; Novack, T.A. Amantadine to improve neurorecovery in traumatic brain injury-associated diffuse axonal injury: A pilot double-blind randomized trial. J. Head Trauma Rehabil. 2002, 17, 300–313. [Google Scholar] [CrossRef]
  23. Schneider, W.N.; Drew-Cates, J.; Wong, T.M.; Dombovy, M.L. Cognitive and behavioural efficacy of amantadine in acute traumatic brain injury: An initial double-blind placebo-controlled study. Brain Inj. 1999, 13, 863–872. [Google Scholar] [CrossRef]
  24. Giacino, J.T.; Whyte, J.; Bagiella, E.; Kalmar, K.; Childs, N.; Khademi, A.; Eifert, B.; Long, D.; Katz, D.I.; Cho, S.; et al. Placebo-controlled trial of amantadine for severe traumatic brain injury. N. Engl. J. Med. 2012, 366, 819–826. [Google Scholar] [CrossRef] [Green Version]
  25. Kraus, M.F.; Smith, G.S.; Butters, M.; Donnell, A.J.; Dixon, E.; Yilong, C.; Marion, D. Effects of the dopaminergic agent and NMDA receptor antagonist amantadine on cognitive function, cerebral glucose metabolism and D2 receptor availability in chronic traumatic brain injury: A study using positron emission tomography (PET). Brain Inj. 2005, 19, 471–479. [Google Scholar] [CrossRef]
  26. Reddy, C.C.; Collins, M.; Lovell, M.; Kontos, A.P. Efficacy of amantadine treatment on symptoms and neurocognitive performance among adolescents following sports-related concussion. J. Head Trauma Rehabil. 2013, 28, 260–265. [Google Scholar] [CrossRef]
  27. Hammond, F.M.; Sherer, M.; Malec, J.F.; Zafonte, R.D.; Dikmen, S.; Bogner, J.; Bell, K.R.; Barber, J.; Temkin, N. Amantadine Did Not Positively Impact Cognition in Chronic Traumatic Brain Injury: A Multi-Site, Randomized, Controlled Trial. J. Neurotrauma 2018, 35, 2298–2305. [Google Scholar] [CrossRef] [Green Version]
  28. Simpson, D.M.; Hallett, M.; Ashman, E.J.; Comella, C.L.; Green, M.W.; Gronseth, G.S.; Armstrong, M.J.; Gloss, D.; Potrebic, S.; Jankovic, J.; et al. Practice guideline update summary: Botulinum neurotoxin for the treatment of blepharospasm, cervical dystonia, adult spasticity, and headache: Report of the Guideline Development Subcommittee of the American Academy of Neurology. Neurology 2016, 86, 1818–1826. [Google Scholar] [CrossRef] [Green Version]
  29. Truong, D.D.; Jost, W.H. Botulinum toxin: Clinical use. Park. Relat. Disord. 2006, 12, 331–355. [Google Scholar] [CrossRef]
  30. Pavesi, G.; Brianti, R.; Medici, D.; Mammi, P.; Mazzucchi, A.; Mancia, D. Botulinum toxin type A in the treatment of upper limb spasticity among patients with traumatic brain injury. J. Neurol. Neurosurg. Psychiatry 1998, 64, 419–420. [Google Scholar] [CrossRef] [Green Version]
  31. Francis, H.P.; Wade, D.T.; Turner-Stokes, L.; Kingswell, R.S.; Dott, C.S.; Coxon, E.A. Does reducing spasticity translate into functional benefit? An exploratory meta-analysis. J. Neurol. Neurosurg. Psychiatry 2004, 75, 1547–1551. [Google Scholar] [CrossRef] [PubMed]
  32. Elia, A.E.; Filippini, G.; Calandrella, D.; Albanese, A. Botulinum neurotoxins for post-stroke spasticity in adults: A systematic review. Mov. Disord. 2009, 24, 801–812. [Google Scholar] [CrossRef] [PubMed]
  33. Foley, N.; Pereira, S.; Salter, K.; Fernandez, M.M.; Speechley, M.; Sequeira, K.; Miller, T.; Teasell, R. Treatment with botulinum toxin improves upper-extremity function post stroke: A systematic review and meta-analysis. Arch. Phys. Med. Rehabil. 2013, 94, 977–989. [Google Scholar] [CrossRef] [PubMed]
  34. Dong, Y.; Wu, T.; Hu, X.; Wang, T. Efficacy and safety of botulinum toxin type A for upper limb spasticity after stroke or traumatic brain injury: A systematic review with meta-analysis and trial sequential analysis. Eur. J. Phys. Rehabil. Med. 2017, 53, 256–267. [Google Scholar] [CrossRef]
  35. Gracies, J.M.; Brashear, A.; Jech, R.; McAllister, P.; Banach, M.; Valkovic, P.; Walker, H.; Marciniak, C.; Deltombe, T.; Skoromets, A.; et al. Safety and efficacy of abobotulinumtoxinA for hemiparesis in adults with upper limb spasticity after stroke or traumatic brain injury: A double-blind randomised controlled trial. Lancet Neurol. 2015, 14, 992–1001. [Google Scholar] [CrossRef]
  36. Dashtipour, K.; Chen, J.J.; Walker, H.W.; Lee, M.Y. Systematic literature review of abobotulinumtoxinA in clinical trials for adult upper limb spasticity. Am. J. Phys. Med. Rehabil. 2015, 94, 229–238. [Google Scholar] [CrossRef] [Green Version]
  37. Jacinto, J.; Varriale, P.; Pain, E.; Lysandropoulos, A.; Esquenazi, A. Patient Perspectives on the Therapeutic Profile of Botulinum Neurotoxin Type A in Spasticity. Front. Neurol. 2020, 11, 388. [Google Scholar] [CrossRef]
  38. Hu, Q.; Manaenko, A.; Xu, T.; Guo, Z.; Tang, J.; Zhang, J.H. Hyperbaric oxygen therapy for traumatic brain injury: Bench-to-bedside. Med. Gas. Res. 2016, 6, 102–110. [Google Scholar] [CrossRef] [Green Version]
  39. Shandley, S.; Wolf, E.G.; Schubert-Kappan, C.M.; Baugh, L.M.; Richards, M.F.; Prye, J.; Arizpe, H.M.; Kalns, J. Increased circulating stem cells and better cognitive performance in traumatic brain injury subjects following hyperbaric oxygen therapy. Undersea Hyperb. Med. 2017, 44, 257–269. [Google Scholar] [CrossRef]
  40. Geng, F.; Ma, Y.; Xing, T.; Zhuang, X.; Zhu, J.; Yao, L. Effects of Hyperbaric Oxygen Therapy on Inflammasome Signaling after Traumatic Brain Injury. Neuroimmunomodulation 2016, 23, 122–129. [Google Scholar] [CrossRef]
  41. Zhong, X.; Shan, A.; Xu, J.; Liang, J.; Long, Y.; Du, B. Hyperbaric oxygen for severe traumatic brain injury: A randomized trial. J. Int. Med. Res. 2020, 48, 300060520939824. [Google Scholar] [CrossRef]
  42. Hadanny, A.; Abbott, S.; Suzin, G.; Bechor, Y.; Efrati, S. Effect of hyperbaric oxygen therapy on chronic neurocognitive deficits of post-traumatic brain injury patients: Retrospective analysis. BMJ Open 2018, 8, e023387. [Google Scholar] [CrossRef] [Green Version]
  43. Leonard, B.E.; McCartan, D.; White, J.; King, D.J. Methylphenidate: A review of its neuropharmacological, neuropsychological and adverse clinical effects. Hum. Psychopharmacol. 2004, 19, 151–180. [Google Scholar] [CrossRef]
  44. Weber, P.; Lutschg, J. Methylphenidate treatment. Pediatr. Neurol. 2002, 26, 261–266. [Google Scholar] [CrossRef]
  45. Sivan, M.; Neumann, V.; Kent, R.; Stroud, A.; Bhakta, B.B. Pharmacotherapy for treatment of attention deficits after non-progressive acquired brain injury. A systematic review. Clin. Rehabil. 2010, 24, 110–121. [Google Scholar] [CrossRef]
  46. Huang, C.H.; Huang, C.C.; Sun, C.K.; Lin, G.H.; Hou, W.H. Methylphenidate on Cognitive Improvement in Patients with Traumatic Brain Injury: A Meta-Analysis. Curr. Neuropharmacol. 2016, 14, 272–281. [Google Scholar] [CrossRef]
  47. Frenette, A.J.; Kanji, S.; Rees, L.; Williamson, D.R.; Perreault, M.M.; Turgeon, A.F.; Bernard, F.; Fergusson, D.A. Efficacy and safety of dopamine agonists in traumatic brain injury: A systematic review of randomized controlled trials. J. Neurotrauma 2012, 29, 1–18. [Google Scholar] [CrossRef] [Green Version]
  48. Iaccarino, M.A.; Philpotts, L.L.; Zafonte, R.; Biederman, J. Stimulant Use in the Management of Mild Traumatic Brain Injury: A Qualitative Literature Review. J. Atten. Disord. 2020, 24, 309–317. [Google Scholar] [CrossRef]
  49. Johansson, B.; Wentzel, A.P.; Andrell, P.; Odenstedt, J.; Mannheimer, C.; Ronnback, L. Evaluation of dosage, safety and effects of methylphenidate on post-traumatic brain injury symptoms with a focus on mental fatigue and pain. Brain Inj. 2014, 28, 304–310. [Google Scholar] [CrossRef]
  50. Johansson, B.; Wentzel, A.P.; Andrell, P.; Mannheimer, C.; Ronnback, L. Methylphenidate reduces mental fatigue and improves processing speed in persons suffered a traumatic brain injury. Brain Inj. 2015, 29, 758–765. [Google Scholar] [CrossRef]
  51. McAllister, T.W.; Zafonte, R.; Jain, S.; Flashman, L.A.; George, M.S.; Grant, G.A.; He, F.; Lohr, J.B.; Andaluz, N.; Summerall, L.; et al. Randomized Placebo-Controlled Trial of Methylphenidate or Galantamine for Persistent Emotional and Cognitive Symptoms Associated with PTSD and/or Traumatic Brain Injury. Neuropsychopharmacology 2016, 41, 1191–1198. [Google Scholar] [CrossRef] [Green Version]
  52. Johansson, B.; Wentzel, A.P.; Andrell, P.; Ronnback, L.; Mannheimer, C. Long-term treatment with methylphenidate for fatigue after traumatic brain injury. Acta Neurol. Scand. 2017, 135, 100–107. [Google Scholar] [CrossRef] [Green Version]
  53. Bickerdike, M.J.; Thomas, G.B.; Batchelor, D.C.; Sirimanne, E.S.; Leong, W.; Lin, H.; Sieg, F.; Wen, J.; Brimble, M.A.; Harris, P.W.; et al. NNZ-2566: A Gly-Pro-Glu analogue with neuroprotective efficacy in a rat model of acute focal stroke. J. Neurol. Sci. 2009, 278, 85–90. [Google Scholar] [CrossRef]
  54. Lu, X.C.; Chen, R.W.; Yao, C.; Wei, H.; Yang, X.; Liao, Z.; Dave, J.R.; Tortella, F.C. NNZ-2566, a glypromate analog, improves functional recovery and attenuates apoptosis and inflammation in a rat model of penetrating ballistic-type brain injury. J. Neurotrauma 2009, 26, 141–154. [Google Scholar] [CrossRef] [PubMed]
  55. Lu, X.C.; Si, Y.; Williams, A.J.; Hartings, J.A.; Gryder, D.; Tortella, F.C. NNZ-2566, a glypromate analog, attenuates brain ischemia-induced non-convulsive seizures in rats. J. Cereb. Blood Flow Metab. 2009, 29, 1924–1932. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Wei, H.H.; Lu, X.C.; Shear, D.A.; Waghray, A.; Yao, C.; Tortella, F.C.; Dave, J.R. NNZ-2566 treatment inhibits neuroinflammation and pro-inflammatory cytokine expression induced by experimental penetrating ballistic-like brain injury in rats. J. NeuroInflamm. 2009, 6, 19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Cartagena, C.M.; Phillips, K.L.; Williams, G.L.; Konopko, M.; Tortella, F.C.; Dave, J.R.; Schmid, K.E. Mechanism of action for NNZ-2566 anti-inflammatory effects following PBBI involves upregulation of immunomodulator ATF3. Neuromol. Med. 2013, 15, 504–514. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Silver, J.M.; Koumaras, B.; Chen, M.; Mirski, D.; Potkin, S.G.; Reyes, P.; Warden, D.; Harvey, P.D.; Arciniegas, D.; Katz, D.I.; et al. Effects of rivastigmine on cognitive function in patients with traumatic brain injury. Neurology 2006, 67, 748–755. [Google Scholar] [CrossRef]
  59. Silver, J.M.; Koumaras, B.; Meng, X.; Potkin, S.G.; Reyes, P.F.; Harvey, P.D.; Katz, D.I.; Gunay, I.; Arciniegas, D.B. Long-term effects of rivastigmine capsules in patients with traumatic brain injury. Brain Inj. 2009, 23, 123–132. [Google Scholar] [CrossRef]
  60. Tenovuo, O.; Alin, J.; Helenius, H. A randomized controlled trial of rivastigmine for chronic sequels of traumatic brain injury-what it showed and taught? Brain Inj. 2009, 23, 548–558. [Google Scholar] [CrossRef]
  61. Brawman-Mintzer, O.; Tang, X.C.; Bizien, M.; Harvey, P.D.; Horner, M.D.; Arciniegas, D.B.; Raskind, M.; Johnson-Greene, L.; Martineau, R.J.; Hamner, M.; et al. Rivastigmine Transdermal Patch Treatment for Moderate to Severe Cognitive Impairment in Veterans with Traumatic Brain Injury (RiVET Study): A Randomized Clinical Trial. J. Neurotrauma 2021, 38, 1943–1952. [Google Scholar] [CrossRef]
  62. Perel, P.; Al-Shahi Salman, R.; Kawahara, T.; Morris, Z.; Prieto-Merino, D.; Roberts, I.; Sandercock, P.; Shakur, H.; Wardlaw, J. CRASH-2 (Clinical Randomisation of an Antifibrinolytic in Significant Haemorrhage) intracranial bleeding study: The effect of tranexamic acid in traumatic brain injury—A nested randomised, placebo-controlled trial. Health Technol. Assess. 2012, 16, 1–54. [Google Scholar] [CrossRef] [Green Version]
  63. Harhangi, B.S.; Kompanje, E.J.; Leebeek, F.W.; Maas, A.I. Coagulation disorders after traumatic brain injury. Acta Neurochir. 2008, 150, 165–175, discussion 175. [Google Scholar] [CrossRef]
  64. Bayir, A.; Kalkan, E.; Kocak, S.; Ak, A.; Cander, B.; Bodur, S. Fibrinolytic markers and neurologic outcome in traumatic brain injury. Neurol. India 2006, 54, 363–365. [Google Scholar] [CrossRef]
  65. Collaborators, C.-T.; Shakur, H.; Roberts, I.; Bautista, R.; Caballero, J.; Coats, T.; Dewan, Y.; El-Sayed, H.; Gogichaishvili, T.; Gupta, S.; et al. Effects of tranexamic acid on death, vascular occlusive events, and blood transfusion in trauma patients with significant haemorrhage (CRASH-2): A randomised, placebo-controlled trial. Lancet 2010, 376, 23–32. [Google Scholar] [CrossRef]
  66. Henry, D.A.; Carless, P.A.; Moxey, A.J.; O’Connell, D.; Stokes, B.J.; McClelland, B.; Laupacis, A.; Fergusson, D. Anti-fibrinolytic use for minimising perioperative allogeneic blood transfusion. Cochrane Database Syst. Rev. 2007, 4, CD001886. [Google Scholar] [CrossRef]
  67. Lawati, K.A.; Sharif, S.; Maqbali, S.A.; Rimawi, H.A.; Petrosoniak, A.; Belley-Cote, E.P.; Sharma, S.V.; Morgenstern, J.; Fernando, S.M.; Owen, J.J.; et al. Efficacy and safety of tranexamic acid in acute traumatic brain injury: A systematic review and meta-analysis of randomized-controlled trials. Intensive Care Med. 2021, 47, 14–27. [Google Scholar] [CrossRef]
  68. Maas, A.I.R.; Steyerberg, E.W.; Citerio, G. Tranexamic acid in traumatic brain injury: Systematic review and meta-analysis trumps a large clinical trial? Intensive Care Med. 2021, 47, 74–76. [Google Scholar] [CrossRef]
  69. Bossers, S.M.; Loer, S.A.; Bloemers, F.W.; Den Hartog, D.; Van Lieshout, E.M.M.; Hoogerwerf, N.; van der Naalt, J.; Absalom, A.R.; Peerdeman, S.M.; Schwarte, L.A.; et al. Association Between Prehospital Tranexamic Acid Administration and Outcomes of Severe Traumatic Brain Injury. JAMA Neurol. 2021, 78, 338–345. [Google Scholar] [CrossRef]
  70. Jelkmann, W. Physiology and pharmacology of erythropoietin. Transfus. Med. Hemother. 2013, 40, 302–309. [Google Scholar] [CrossRef] [Green Version]
  71. Bramlet, H.M.; Dietrich, W.D.; Dixosn, C.E.; Shear, D.A.; Schmid, K.E.; Modello, S.; Wang, K.K.; Hayes, R.L.; Povlishock, J.T.; Tortella, F.C.; et al. Erythropoietin treatment in trauamtic brain injury: Operation brain trauma therapy. J. Neurotrauma 2016, 33, 538–552. [Google Scholar] [CrossRef]
  72. Cherian, L.; Goodman, J.C.; Robertson, C. Neuroprotection with erythropoietin administration following controlled cortical impact injury in rats. J. Pharm. Exp. 2007, 322, 789–794. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Peng, W.; Xing, Z.; Yang, J.; Wang, Y.; Wang, W.; Huang, W. The efficacy of erythropoietin in treating experimental traumatic brain injury: A systematic review of controlled trials in animal models. J. Neurosurg. 2014, 121, 653–664. [Google Scholar] [CrossRef] [Green Version]
  74. Kabadi, S.V.; Faden, A.I. Neuroprotective strategies for traumatic brain injury: Improving clinical translation. Int. J. Mol. Sci. 2014, 15, 1216–1236. [Google Scholar] [CrossRef] [Green Version]
  75. Grasso, G.; Alafaci, C.; Buemi, M. Erythropoietin in traumatic brain injury: An answer will come soon. World Neurosurg. 2015, 84, 1491–1492. [Google Scholar] [CrossRef]
  76. Liu, W.C.; Wen, L.; Xie, T.; Wang, H.; Gong, J.B.; Yang, X.F. Therapeutic effect of erythropoietin in patients with traumatic brain injury: A meta-analysis of randomized controlled trials. J. Neurosurg. 2016, 127, 8–15. [Google Scholar] [CrossRef]
  77. Liu, M.; Wang, A.J.; Chen, C.; Zhao, G.; Jiang, Z.; Wang, X.; Shi, D.; Zhang, T.; Sun, B.; He, S.; et al. Efficacy and safety of erythropoietin for traumatic brain injury. BMC Neurol. 2020, 20, 399. [Google Scholar] [CrossRef]
  78. Temkin, N.R.; Dikmen, S.S.; Wilensky, A.J.; Keihm, J.; Chabal, S.; Winn, R. A randomized, double-blind study of phenytoin for the prevention of post-traumatic seizures. N. Engl. J. Med. 1990, 323, 497–502. [Google Scholar] [CrossRef]
  79. Khan, S.A.; Bhatti, S.N.; Khan, A.A.; Afridi, E.A.K.; Muhammad, G.; Gul, N.; Zadran, K.K.; Alam, S.; Aurangzeb, A. Comparison of efficacy of Phenytoin and Levetiracetam for prevention of early post traumatic seizures. J. Ayub. Med. Coll. Abbottabad 2016, 28, 455–460. [Google Scholar] [PubMed]
  80. McGinn, R.J.; Aljoghaiman, M.S.; Sharma, S.V. Levetiracetam vs phenytoin prophylaxis in severe traumatic brain injury: Systematic review and meta-analysis. Interdiscip. Neurosurg. 2022, 27, 101394. [Google Scholar] [CrossRef]
  81. Spyropoulos, A.C.; Hussein, M.; Lin, J.; Battleman, D. Rates of venous thromboembolism occurrence in medical patients among the insured population. Thromb. Haemost. 2009, 102, 951–957. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Schaible, E.-V.; Thal, S.C. Anticoagulation in patients with traumatic brain injury. Curr. Opin. Anaesthesiol. 2013, 26, 529–534. [Google Scholar] [CrossRef] [PubMed]
  83. Dudley, R.R.; Azi, I.; Bonnici, A.; Saluja, R.S.; Lamoureux, J.; Kalmovitch, B.; Gursahaney, A.; Razek, T. Early Venous Thromboembolic Event Prophylaxis in Traumatic Brain Injury with Low-Molecular-Weight Heparin: Risks and Benefits. J. Neurotrauma 2010, 27, 12. [Google Scholar] [CrossRef]
  84. 8Margilick, J.; Dandurand, C.; Duncan, K.; Chen, W.; Evans, D.C.; Sekhon, M.S.; Garraway, N.; Griesdale, D.E.G.; Gooderham, P.; Hameed, S.M. A Systematic Review of the Risks and Benefits of Venous Thromboembolism Prophylaxis in Traumatic Brain Injury. Can. J. Neurol. Sci. 2018, 45, 432–444. [Google Scholar] [CrossRef]
  85. Gunning, A.C.; Maier, R.V.; de Rooij, D.; de Leenen, D.; Leenen, L.P.H.; Hietbrink, F. Venous thromboembolism (VTE) prophylaxis in severely injured patients: An international comparative assessment. Eur. J. Trauma Emerg. Surg. 2021, 47, 137–143. [Google Scholar] [CrossRef] [Green Version]
  86. Woolf, P.D.; Hamill, R.W.; Lee, L.A.; Cox, C.; McDonald, J.V. The predictive value of catecholamines in assessing outcome in traumatic brain injury. J. Neurosurg. 1987, 66, 875–882. [Google Scholar] [CrossRef] [Green Version]
  87. Woolf, P.D.; McDonald, J.V.; Feliciano, D.V.; Kelly, M.M.; Nichols, D.; Cox, C. The catecholamine response to multisystem trauma. Arch. Surg. 1992, 127, 899–903. [Google Scholar] [CrossRef]
  88. Perkes, I.; Baguley, I.J.; Nott, M.T.; Menon, D.K. A review of paroxysmal sympathetic hyperactivity after acquired brain injury. Ann. Neurol. 2010, 68, 126–135. [Google Scholar] [CrossRef]
  89. Riordan, W.P.; Cotton, B.A.; Norris, P.R.; Waitman, L.R.; Jenkins, J.M.; Morris, J.A. Beta-blocker exposure in patients with severe traumatic brain injury (TBI) and cardiac uncoupling. J. Trauma 2007, 63, 503–510. [Google Scholar] [CrossRef]
  90. Nott, M.T.; Chapparo, C.; Baguley, I.J. Agitation following traumatic brain injury: An Australian sample. Brain Inj. 2006, 20, 1175–1182. [Google Scholar] [CrossRef]
  91. Baguley, I.J.; Slewa-Younan, S.; Heriseanu, R.E.; Nott, M.T.; Mudaliar, Y.; Nayyar, V. The incidence of dysautonomia and its relationship with autonomic arousal following traumatic brain injury. Brain Inj. 2007, 21, 1175–1181. [Google Scholar] [CrossRef]
  92. Liu, M.Y. Protective effects of propranolol on experimentally head-injured mouse brains. J. Med. Assoc. 1995, 94, 386–390. [Google Scholar]
  93. Ley, E.J.; Park, R.; Dagliyan, G.; Palestrant, D.; Miller, C.M.; Conti, P.S.; Margulies, D.R.; Salim, A. In vivo effect of propranolol dose and timing on cerebral perfusion after traumatic brain injury. J. Trauma 2010, 68, 353–356. [Google Scholar] [CrossRef]
  94. Ley, E.J.; Scehnet, J.; Park, R.; Schroff, S.; Dagliyan, G.; Conti, P.S.; Margulies, D.R.; Salim, A. The in vivo effect of propranolol on cerebral perfusion and hypoxia after traumatic brain injury. J. Trauma 2009, 66, 154–159. [Google Scholar] [CrossRef]
  95. Brooke, M.M.; Patterson, D.R.; Questad, K.A.; Cardenas, D.; Farrel-Roberts, L. The treatment of agitation during initial hospitalization after traumatic brain injury. Arch. Phys. Med. Rehabil. 1992, 73, 917–921. [Google Scholar]
  96. Fleminger, S.; Greenwood, R.J.; Oliver, D.L. Pharmacological management for agitation and aggression in people with acquired brain injury. Cochrane Database Syst. Rev. 2006, 4, CD003299. [Google Scholar] [CrossRef]
  97. Hinson, H.E.; Sheth, K.N. Manifestations of the hyperadrenergic state after acute brain injury. Curr. Opin. Crit. Care 2012, 18, 139–145. [Google Scholar] [CrossRef]
  98. Tran, T.Y.; Dunne, I.E.; German, J.W. Beta blockers exposure and traumatic brain injury: A literature review. Neurosurg. Focus 2008, 25, E8. [Google Scholar] [CrossRef] [Green Version]
  99. Gomes, J.A.; Stevens, R.D.; Lewin, J.J.; Misrki, M.A.; Bhardwaj, A. Glucocorticoid therapy in neurologic critical care. Crit. Care Med. 2005, 33, 1214–1224. [Google Scholar] [CrossRef]
  100. Moll, A.; Lara, M.; Pomar, J.; Orozco, M.; Frontera, G.; Llompart-Pou, J.A.; Moratinos, L.; Gonzalez, V.; Ibanez, J.; Perez-Barcena, J. Effects of dexamethasone in traumatic brain injury patients with pericontusional vasogenic edema: A prospective-observational DTI-MRI study. Medicine 2020, 23, e22879. [Google Scholar] [CrossRef]
  101. Roberts, I.; Yates, D.; Sandercock, P.; Farrell, B.; Wasserberg, J.; Lomas, G. Effect of intravenous corticosteroids on death within 14 days in 10008 adults with clinically significant head injury (MRC CRASH trial): Randomised placebo-controlled trial. Lancet 2004, 364, 1318–1321. [Google Scholar] [CrossRef] [PubMed]
  102. The CRASH-3 Collaborators. Effects of tranexamic acid on death, disability, vascular occlusive events and other morbidities in patients with acute traumatic brain injury (CRASH-3): A randomised, placebo-controlled trial. Lancet 2019, 394, 1713–1723. [Google Scholar] [CrossRef] [Green Version]
  103. Crash-3 Trial Results. Available online: https://crash3.lshtm.ac.uk/blog/crash-3-trial-results/ (accessed on 13 April 2022).
  104. Sharma, S.; Verma, S. Is positive publication bias really a bias, or an intentionally created discrimination toward negative results? Saudi J. Anaesth. 2019, 13, 352–355. [Google Scholar] [CrossRef]
  105. Mlinaric, A.; Horvat, M.; Smolcic, V.S. Dealing with the positive publication bias: Why you should really publish your negative results. Biomech. Med. 2017, 27, 030201. [Google Scholar] [CrossRef] [Green Version]
  106. The importance of no evidence. Nat. Hum. Behav. 2019, 3, 197. [CrossRef]
  107. Bespalov, A.; Steckler, T.; Skolnick, P. Be positive about negatives–recommendations for the publication of negative (or null) results. Eur. Neuropharmacol. 2019, 29, 1312–1320. [Google Scholar] [CrossRef]
  108. Kochanek, P.M.; Jackson, T.C.; Jha, R.M.; Clark, R.S.B.; Okonkwo, D.O.; Bayir, H.; Poloyac, S.M.; Wagner, A.K.; Empey, P.E.; Conley, Y.P.; et al. Paths to Successful Translation of New Therapies for Severe Traumatic Brain Injury in the Golden Age of Traumatic Brain Injury Research: A Pittsburgh Vision. J. Neurotrauma 2020, 37, 2353–2371. [Google Scholar] [CrossRef]
  109. Huijben, J.A.; Volovici, V.; Cnossen, M.C.; Haitsma, I.K.; Stocchetti, N.; Maas, A.I.R.; Menon, D.K.; Ercole, A.; Citerio, G.; Nelson, D.; et al. Variation in general supportive and preventive intensive care management of traumatic brain injury: A survey in 66 neurotrauma centers participating in the Collaborative European NeuroTrauma Effectiveness Research in Traumatic Brain Injury (CENTER-TBI) study. Crit. Care 2018, 22, 90. [Google Scholar] [CrossRef] [Green Version]
  110. Kurz, J.E.; Poloyac, S.M.; Abend, N.S.; Fabio, A.; Bell, M.J.; Wainwright, M.S. Variation in Anticonvulsant Selection and Electroencephalographic Monitoring Following Severe Traumatic Brain Injury in Children-Understanding Resource Availability in Sites Participating in a Comparative Effectiveness Study. Pediatr. Crit. Care Med. 2016, 17, 649–657. [Google Scholar] [CrossRef] [PubMed]
  111. Talisa, V.B.; Yende, S.; Seymour, C.W.; Angus, D.C. Arguing for Adaptive Clinical Trials in Sepsis. Front. Immunol. 2018, 9, 1502. [Google Scholar] [CrossRef]
  112. Shein, S.L.; Ferguson, N.M.; Kochanek, P.M.; Bayir, H.; Clark, R.S.; Fink, E.L.; Tyler-Kabara, E.C.; Wisniewski, S.R.; Tian, Y.; Balasubramani, G.K.; et al. Effectiveness of Pharmacological Therapies for Intracranial Hypertension in Children With Severe Traumatic Brain Injury—Results from an Automated Data Collection System Time-Synched to Drug Administration. Pediatr. Crit. Care Med. 2016, 17, 236–245. [Google Scholar] [CrossRef]
  113. Young, A.M.; Donnelly, J.; Czosnyka, M.; Jalloh, I.; Liu, X.; Aries, M.J.; Fernandes, H.M.; Garnett, M.R.; Smielewski, P.; Hutchinson, P.J.; et al. Continuous Multimodality Monitoring in Children after Traumatic Brain Injury-Preliminary Experience. PLoS ONE 2016, 11, e0148817. [Google Scholar] [CrossRef]
Figure 1. Status of clinical trials in TBI listed on ClinicalTrials.gov as of 3 December 2021.
Figure 1. Status of clinical trials in TBI listed on ClinicalTrials.gov as of 3 December 2021.
Brainsci 12 00527 g001
Figure 2. Number of completed clinical trials in TBI in each year from 2002–2021.
Figure 2. Number of completed clinical trials in TBI in each year from 2002–2021.
Brainsci 12 00527 g002
Figure 3. Number of participants for clinical trials in TBI and their age groupings in the completed studies. Note: y axis breaks at 500 participants.
Figure 3. Number of participants for clinical trials in TBI and their age groupings in the completed studies. Note: y axis breaks at 500 participants.
Brainsci 12 00527 g003
Figure 4. Completed clinical trials in TBI and their study phase/design.
Figure 4. Completed clinical trials in TBI and their study phase/design.
Brainsci 12 00527 g004
Figure 5. Clinical trials of drugs appearing in three or more registered clinical trials.
Figure 5. Clinical trials of drugs appearing in three or more registered clinical trials.
Brainsci 12 00527 g005
Table 1. Completed studies with results summarized.
Table 1. Completed studies with results summarized.
NCT NumberInterventionAge (Years)EnrollmentPrimary OutcomeSummary of Results
NCT01990768Tranexamic acid15 and older388Dichotomized Glasgow Outcome Scale Extended (GOS-E) at 6 monthsNo statistical difference between 1 g IV TXA followed by 1 g IV maintenance or 2 g TXA IV TXA only and placebo: 967 patients enrolled.
NCT00313716Recombinant human erythropoietin15 and older200Glasgow Outcome Scale at 6 monthsNo statistical differences between Epo treatment and placebo. All 200 patients enrolled. Results published.
NCT01322048IV Propranolol vs. clonidine16 to 6463Ventilator-free daysNo statistical change in ventilator-free days using Propanolol and clonidine. Secondary outcomes of plasma norepinephrine levels showed higher levels after treatment: 48 patients enrolled. Results published.
NCT01673828Allopregnanolone injection16 to 65967Dichotomized Glasgow Outcome Scale Extended (GOS-E) at 6 monthsNo statistical difference between treatment and placebo in primary outcomes. Only 13 participants enrolled.
NCT00970944Amantadine Hydrochloride16 to 65200Disability rating score (DRS)No statistical difference in DRS scores over 6 weeks between treated and placebo. However, DRS score improved faster in the amantadine group in the first 4 weeks, then significantly slower than the normal group: 184 patients enrolled.
NCT01201863Androgel16 to 6541Restricted functional independence measure (FIM)No statistical difference between treated and placebo: 46 patients enrolled.
NCT00621751Carbamazepine16 to 65157Neuropsychiatric Inventory Irritability-Aggression Domains Composite Measure—ObserverNo statistical difference between treated and placebo: 70 patients enrolled.
NCT00779324Amantadine Hydrochloride16 to 7552Proportion of Participants With >2-point increase on Neuropsychiatric Inventory—Irritability Domain rated by Observer day 28No statistical difference between treated and placebo: 168 patients enrolled.
NCT02957331Propranolol18 and older6330-day mortalityIn the treatment arm, 7.7% of patients died, whilst 33.3% of patients died in the non-treatment arm. No statistical data present: 26 patients enrolled.
NCT01014403Enoxaparin18 and older184Percentage of participants with worsening TBI hemorrhageNo statistical difference between treated and placebo: 62 patients enrolled.
NCT01847755Oxygen at 1.5 ATA18 and older71Number of participants with improved cerebral perfusionSingle group allocation and so no comparisons possible: 18 patients enrolled.
NCT00795366Arginine vasopressin vs. catecholamine18 and older60Time ICP > 20No statistical difference between treated and standard catecholamine: 96 patients enrolled.
NCT00618436Levetiracetam vs. phenytoin18 and older47Seizure incidenceNo statistical difference between Levetiracetam or phenytoin: 52 patients enrolled.
NCT00233103Sertraline18 and older30Depression at baselineNo statistical difference between treatment and placebo: 52 patients enrolled.
NCT01368432Escitalopram18 and older30Montgomery–Asberg depression rating scale (MADRS) at baselineNo statistical difference between treatment and placebo: 16 patients enrolled.
NCT00766038Recombinant human growth hormone18 to 50336Functional outcome 6 months after injury, as measured by the processing speed indexNo statistical difference between treatment and placebo: 63 patients enrolled.
NCT00973674Premarin IV18 to 50256Percent passing the Galveston orientation amnesia test (GOAT) within 28 days post injuryNo statistical difference between treatment and placebo: 50 patients enrolled.
NCT00623506Pregnenolone18 to 5550Brief assessment of cognition in affective disorders (BAC-A)No statistical difference between treatment and placebo: 30 patients enrolled.
NCT02225106Methylphenidate18 to 5534Perceptual organization and processing speed indexSingle group allocation and so no comparisons possible: 11 patients enrolled.
NCT00727246CDP-Choline18 to 5513Cognitive composite score for group of subjects with TBI and healthy controls matched by age, education and treatment groupNo statistical difference between treatment and placebo. Low group numbers: 19 patients enrolled.
NCT01856270Amitriptyline18 to 6012,737Frequency and severity of headachesNo statistical difference between treatment and placebo: 50 patients enrolled.
NCT01342549Valproate and Naltrexone18 to 6015Time to relapse to heavy drinkingPatients on Naltrexone relapsed to heavy drinking at 15.99 weeks compared to those on Valproate who relapsed after 8.78 weeks: 62 patients enrolled.
NCT02025439Amantadine18 to 6441Intensity of adverse eventPatients receiving repetitive transcranial magnetic stimulation (rTMS) combined with amantadine (TMS + amantadine) had significantly higher intensity of adverse events. Patient numbers very low: 4 patients enrolled.
NCT01760785Divalproex sodium18 to 65606Severity of affective lability based on shortened agitated behavior scaleNo statistical difference between treatment and placebo: 50 patients enrolled.
NCT02240589Memantine18 to 65238California Verbal Learning Test—Second Edition (CVLT-II)—Long Delay Free RecallMemantine group had a higher z-score (−2.000), i.e., worse performance, compared to placebo (−1.375): 11 patients enrolled.
NCT00702364Atomoxetine18 to 65179CDR power of attention and Stroop test interference T-scoreNo statistical differences between treatment and placebo: 60 patients enrolled.
NCT02012582VAS20318 to 65100Effects on intracranial pressure (ICP), cerebral perfusion pressure (CPP), brain metabolism using microdialysis and the therapy intensity level (TIL). Extended Glasgow outcome score (eGOS) at 6 months was exploratoryNo statistical differences between treatment and placebo for ICP, CPP and brain metabolism. Scores for eGOS were significantly higher in treated groups: 32 patients enrolled. Results published.
NCT01750268Topiramate18 to 6560Change in the number of drinking days per week as assessed by the timeline followback (TLFB)No statistical difference between treatment arm and placebo. Topiramate transiently impaired verbal fluency and working memory. No statistical difference in processing speed, cognitive inhibition and mental flexibility: 32 patients enrolled. Results published.
NCT01611194Hyperbaric oxygen18 to 6550Summary of treatment-emergent adverse eventsHigher incidence of Barotitis media, upper respiratory tract infection and eye disorders in treated group compared to control groups: 71 patients enrolled.
NCT01306968Hyperbaric oxygen18 to 6547Post-intervention post-concussion symptom scores using RPQNo statistical differences between treatment and placebo: 79 patients enrolled.
NCT02791945N-acetylcysteine18 to 6532Change in percent of heavy drinking days per week as assessed by the timeline followback (TFLB)No statistical differences between treatment and placebo: 30 patients enrolled.
NCT00453921Methylphenidate18 to 6531Neuropsychological assessment, CVLT-II and CPT, distractibility condition (reaction time)No statistical differences between treatment and placebo in CVLT-II or CPT: 76 patients enrolled.
NCT01854385Sumatriptan18 to 6516Change in headache reliefSingle arm study so comparisons not possible: 40 patients enrolled.
NCT01249404Botulinum toxin type A18 to 8018Least squares mean change from baseline to week 4 in the mas score in the gastrocnemius-soleus complex (GSC) (knee extended)No statistical differences between treatment and placebo at 1000 U of Dysport, but statistically significant at 1500 U Dysport (p = 0.0091): 388 patients enrolled.
NCT01313299Botulinum toxin type A18 to 8016Change from baseline in mas score in the primary targeted muscle group (PTMG)Treatment 500 and 1000 U of Dysport caused worse outcomes compared to placebo groups: 243 patients enrolled.
NCT00704379Sertraline18 to 85200Time to onset of diagnostic and statistical manual (DSM) IV defined mood and anxiety disorders associated with TBIDSM appeared significantly earlier in treated groups (mean 15.78 weeks) compared to 21.42 weeks in placebo treated controls (p < 0.05): 94 patients enrolled.
NCT01670526Rivastigmine transdermal patch19 to 6540Improvements from baseline on the Hopkins verbal learning test-revised (HVLT-R) total recallNo statistical differences between treatment and placebo: 94 patients enrolled.
NCT02270736NT 2012 to 1717Change from baseline in unstimulated salivary flow rate (uSFR) at week 4Superiority of NT201 shown with a significant uSFR decrease with NT201 compared to placebo (p = 0.0012). Secondary outcome of Carer’s GICS rating was also significant and favored treatment (p = 0.032): 256 patients enrolled. Results published.
NCT01322009Probenecid and N-acetyl cysteine2 to 1817Number of participants who experienced adverse eventsNone in the treatment arm experienced adverse events; 2 patients in the placebo arm experienced adverse events: 14 patients enrolled.
NCT00957671Recombinant human growth hormone21 and older210Maximum oxygen uptake at baseline and maximum oxygen uptake after one yearSingle arm study so comparisons not possible: 15 patients enrolled.
NCT01336413Pregnenolone21 to 5534CAPS (Cluster D symptoms)—primary behavioral outcome measure and Tower of London (Subscale Test of BAC)—primary cognitive outcome measureNo significant differences in CAPS between treatment and placebo. Tower of London score slightly better at 4 weeks but worse at 8 weeks compared to placebo: 53 patients enrolled.
NCT01463033Levetiracetam6 and older15Post-traumatic epilepsyNo significant difference in post-traumatic epilepsy rate in treated versus control arms: 126 patients enrolled. Results published.
NCT02712996Lisdexamfetamine6 to 16150Assessing severity of symptoms associated with attention-deficit/hyperactivity disorder (ADHD) using the Conners-3 parent form and executive using the behavior rating inventory of executive function (BRIEF)Better overall scores in the Conners-3 and BRIEF outcomes. No statistical data present: 20 patients enrolled.
NCT01933217Methylphenidate6 to 17300Parent outcome-Vanderbilt ADHD parent rating scales (VADPRS) and parent outcome-behavior rating inventory of executive functioning (BRIEF)Slightly better overall scores in VADPRS and BRIEF outcomes. No statistical data present: 26 patients enrolled.
Table 2. Studies active but not recruiting (as of 3 December 2021).
Table 2. Studies active but not recruiting (as of 3 December 2021).
NCT NumberInterventionAge (Years)EnrollmentPhase of TrialDate Last Updated
NCT04833218Propanolol18 to 6090Early Phase I6 April 2021
NCT03554265Somatropin18 to 7054Phase III30 November 2021
NCT02255799Donepezil18 to 60160Phase III26 April 2021
Table 3. Studies active and currently recruiting (as of 3 December 2021).
Table 3. Studies active and currently recruiting (as of 3 December 2021).
NCT NumberInterventionAge (Years)EnrollmentPhase of TrialDate Last Updated
NCT04487275MLC901 15 to 6580Phase IV27 July 2020
NCT05033444PRV-00218 to 5524Phase I24 September 2021
NCT01048138Biperiden Lactate18 to 75132Phase III18 June 2021
NCT02404779Cisatracurium besilate18 and older34Phase IV7 July 2020
NCT04550377Cannabidiol18 to 70120Phase II2 June 2021
NCT04303065Dexamethasone18 to 85600Phase III23 November 2020
NCT03559114Dalteparin18 and older1100Phase III15 June 2021
NCT04489160C1 Inhibitor18 to 64106Phase II13 September 2021
NCT04006054Dexmedetomidine vs. Midazolam18 to 7082Phase IV2 July 2019
NCT01821690Buspirone18 to 7074NA2 April 2021
NCT03061565Erythropoietin15 to 75603Not specified25 August 2021
NCT03992404NT 20118 to 85600Phase III22 November 2021
NCT04521881Tranexamic acid50 and older10,000Phase III20 October 2021
NCT02990091Omega 3 fatty acid/Safflower seed oil18 to 5545Phase II23 September 2021
NCT04974060Remifentanil injection18 and older30NA29 July 2021
NCT04280965Quetiapine Fumarate18 and older20Early Phase I1 March 2021
NCT03982602Ketogenic diet18 and older10Early Phase I12 October 2021
NCT03260569Inhaled nitric oxide18 and older38Phase III3 November 2020
NCT04527289Amantadine18 to 7550Phase IV5 October 2021
NCT04426487Progesterone20 to 65200Early Phase I11 June 2020
NCT04508244Propranolol18 to 65771Phase IV21 February 2021
NCT04718155Atorvastatin18 to 4030NA25 March 2021
NCT04244058Amantadine + L-DOPA18 to 7530Early Phase I4 October 2021
NCT05097261Ketamine18 and older100Phase IV28 October 2021
NCT03095066AVP-78618 to 75150Phase II29 November 2021
NCT04499755Nucleo CMP Forteup to 18100Phase III5 August 2020
NCT04558346Ghrelin (OXE-103)18 to 6040Phase II28 July 2021
NCT03417492Sildenafil Citrate40 to 6530Phase I23 September 2021
NCT04673240Botulinum toxin type A injection18 and older70Not specified17 December 2020
NCT02407028Hyperbaric oxygen16 to 65 Years200Phase II27 August 2021
NCT03814356Methylphenidate18 and older22Phase I2 July 2021
NCT04117672Salovum (dietary supplement)10 to 7020Phase II24 March 2021
NCT04588311Epoetin Alfa 40000 UNT/ML18 to 752500Phase III25 August 2021
NCT04631484Cytoflavin18 to 60320Phase III18 August 2021
NCT04099667Rimabotulinumtoxin B18 to 80272Phase II/III15 July 2021
NCT04710550F18-3F4AP18 to 9066Phase I24 February 2021
NCT04744051Adipose Derived Stem Cell Infusion18 to 6520Phase I9 February 2021
Table 4. Studies active but not recruiting (as of 3 December 2021).
Table 4. Studies active but not recruiting (as of 3 December 2021).
NCT NumberInterventionAge (Years)EnrollmentPhase of TrialDate Last Updated
NCT05049057Erenumab18 to 50404Phase II17 September 2021
NCT04573803Phenytoin Sodium10 and older1649Phase III3 November 2020
NCT04945213Biperiden18 to 75312Phase III22 October 2021
NCT04003285Allopregnanolone21 to 62132Phase II23 September 2021
NCT05058677Aerosolized 2% lidocaineup to 1612Phase IV11 October 2021
NCT04400266B + MEL18 to 6410Phase IV22 May 2020
NCT04427241Amantadine Sulfate + Cerebrolysin19 to 6412Phase IV11 June 2020
NCT04867317Somatropin21 to 55172Phase III15 October 2021
NCT05095857S-ketamine18 and older400Phase IV27 October 2021
NCT04815967Rimabotulinumtoxin B18 to 80272Phase II/III15 July 2021
NCT05008926Naloxegol18 and older370Phase III23 August 2021
NCT04387305Tranexamic acid injectionup to 172000Phase III12 July 2021
NCT04515420Noradrenaline18 to 8060Not specified17 August 2020
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ahmed, Z. Current Clinical Trials in Traumatic Brain Injury. Brain Sci. 2022, 12, 527. https://doi.org/10.3390/brainsci12050527

AMA Style

Ahmed Z. Current Clinical Trials in Traumatic Brain Injury. Brain Sciences. 2022; 12(5):527. https://doi.org/10.3390/brainsci12050527

Chicago/Turabian Style

Ahmed, Zubair. 2022. "Current Clinical Trials in Traumatic Brain Injury" Brain Sciences 12, no. 5: 527. https://doi.org/10.3390/brainsci12050527

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