Head Injury

Back

Practice Essentials

Head injury can be defined as any alteration in mental or physical functioning related to a blow to the head (see the image below). According to the Centers for Disease Control and Prevention (CDC), more than 50,000 individuals die from traumatic brain injuries each year in the United States. Almost twice that many people suffer permanent disability. In the United States in 2013, about 2.8 million emergency department (ED) visits, hospitalizations, or deaths were associated with TBI—either alone or in combination with other injuries.[1, 2]



View Image

This 50-year-old woman with epilepsy seized and struck her head. Her initial Glasgow Coma Scale score was 12. Her scan shows prominent right temporal ....

Signs and symptoms

The Glasgow Coma Scale (GCS) is the mainstay for rapid neurologic assessment in acute head injury. Following ascertainment of the GCS score, the examination is focused on signs of external trauma, as follows:

See Clinical Presentation for more detail.

Diagnosis

Bedside cognitive testing

In the acute setting, measurements of the patient's level of consciousness, attention, and orientation are of primary importance.

Some patients acutely recovering from head trauma demonstrate no ability to retain new information. Mental status assessments have validated the prognostic value of the duration of posttraumatic amnesia; patients with longer durations of posttraumatic amnesia have poorer outcomes.[7]

In the long-term setting, the following bedside cognitive tests can be employed:

Laboratory studies

Imaging studies

Electroencephalography

Although certain electroencephalographic patterns may have prognostic significance, considerable interpretation is needed, and sedative medications and electrical artifacts are confounding. The most useful role of electroencephalography (EEG) in head injuries may be to assist in the diagnosis of nonconvulsive status epilepticus.

See Workup for more detail.

Management

Intracranial pressure

If the intracranial pressure rises above 20-25 mm Hg, intravenous mannitol, cerebrospinal fluid drainage, and hyperventilation can be used. If the intracranial pressure does not respond to these conventional treatments, high-dose barbiturate therapy is permissible.[13]

Another approach used by some clinicians is to focus primarily on improving cerebral perfusion pressure as opposed to intracranial pressure in isolation.

Decompressive craniectomies are sometimes advocated for patients with increased intracranial pressure refractory to conventional medical treatment.

Hypertonicity

Dantrolene, baclofen, diazepam, and tizanidine are current oral medication approaches to hypertonicity. Baclofen and tizanidine are customarily preferred because of their more favorable side-effect profiles.

Subdural hematomas

Traditionally, the prompt surgical evacuation of subdural hematomas was believed to be a major determinant of an optimal outcome. However, research indicates that the extent of the original intracranial injury and the generated intracranial pressures may be more important than the timing of surgery.

See Treatment and Medication for more detail.

Background

Head injury can be defined as any alteration in mental or physical functioning related to a blow to the head. Loss of consciousness does not need to occur. The severity of head injuries is most commonly classified by the initial postresuscitation Glasgow Coma Scale (GCS) score, which generates a numerical summed score for eye, motor, and verbal abilities. Traditionally, a score of 13–15 indicates mild injury, a score of 9-12 indicates moderate injury, and a score of 8 or less indicates severe injury. In the last few years, however, some studies have included those patients with scores of 13 in the moderate category, while only those patients with scores of 14 or 15 have been included as mild.[14] Concussion and mild head injury are generally synonymous.

Research on head injury has advanced considerably in the last decade. As is typical of many endeavors, these efforts have exposed the complexity of this condition more deeply and have helped researchers and physicians to abandon crude simplifications. This review concentrates primarily on current developments in the diagnosis and management of closed head injuries in adults.

Pathophysiology

Structural changes

Gross structural changes in head injury are common and often obvious both on autopsy and conventional neuroimaging. The skull can fracture in a simple linear fashion or in a more complicated depressed manner, in which bone fragments and pushes beneath the calvarial surface. In patients with mild head injury, a skull fracture markedly increases the chance of significant intracranial injury.

Both direct impact and contrecoup injuries, in which the moving brain careens onto the distant skull opposite the point of impact, can result in focal bleeding beneath the calvaria. Such bleeding can result in an intracerebral focal contusion or hemorrhage as well as an extracerebral hemorrhage. Extracerebral hemorrhages are primarily subdural hemorrhages arising from tearing of bridging veins, but epidural hemorrhages from tearing of the middle meningeal artery or the diploic veins are also common. Occasionally, subdural hemorrhages can result from disruption of cortical arteries. This type of subdural hemorrhage is rapidly progressive and can occur after trivial head injury in elderly patients.[15]

One study of CT images from 753 patients with severe head injury from the National Institute of Health Traumatic Coma Data Bank in the United States found evidence of intracranial hemorrhagic lesions in 27%. Traumatic subarachnoid hemorrhage was even more frequent and occurred in 39% of patients. Furthermore, diffuse cerebral edema also was present in 39%. Cerebral edema can be unilateral or diffuse and can occur even in the absence of intracranial bleeding. Severe brain edema probably occurs more commonly in children than in adults.[16]

Neuronal loss is also important. A recent pathological study found that quantitative loss of neurons from the dorsal thalamus correlated with severe disability and vegetative state outcomes in patients with closed head injuries.[17]

Finally, axonal injury increasingly has been recognized as a structural sequela of brain injury. The use of amyloid precursor protein staining has resulted in increased recognition of this form of injury. Using this technique, researchers have readily identified axonal injury in patients with mild head injury. Interestingly, a prominent locus of axonal damage has been the fornices, which are important for memory and cognition.[18] More severe and diffuse axonal injury has been found to correlate with vegetative states and the acute onset of coma following injury.[19]

Neurochemical changes

After traumatic brain injury, the brain is bathed with potentially toxic neurochemicals. Catecholamine surges have been documented in the plasma (higher catecholamine levels correlated with worse clinical outcomes) and in the cerebrospinal fluid (CSF) of patients with head injuries (higher CSF 5-hydroxyindole acetic acid (HIAA), the serotonin metabolite, correlated with worse outcomes).[20]  In addition, the excitotoxic amino acids (ie, glutamate, aspartate) initiate a cascade of processes culminating in an increase in intraneuronal calcium and cell death. Researchers using a microdialysis technique have correlated high CSF levels of excitotoxic amino acids with poor outcomes in head injury.[21]

Although neuroprotective strategies employing antiexcitotoxic pharmacotherapies were effective in diminishing the effects of experimental brain injuries in laboratory animals, clinical trials in humans generally have been disappointing.[22] These failures have prompted development of more complex models of neuronal injury and cell death. Recently, researchers have demonstrated that although certain types of glutamate antagonists may protect against acute cell death, they potentiate slowly progressive neuronal injury in experimental rodent models. Still others have found that low-dose glutamate administered before brain injury is somehow neuroprotective. Such dose and timing effects are only beginning to be understood.[23]

Prostaglandins, inflammatory mediators produced by membrane lipid breakdown, are also elevated dramatically in the plasma of patients with moderate-to-severe head trauma during the first 2 weeks after injury. Patients with higher prostaglandin levels had significantly worse outcomes than those with more modest elevations. Furthermore, levels of a thromboxane metabolite, a potent vasoconstricting prostaglandin, were elevated disproportionately.[24] Such a process may underlie posttraumatic vasospasm, which has been documented in some, but not all, transcranial Doppler studies of patients with closed head injuries, even in patients without traumatic subarachnoid bleeds.[25]  Delayed clinical deterioration could represent ischemia from such vasospasm, particularly in younger patients.[26]

Head injury also causes the release of free radicals and the breakdown of membrane lipids. Panels of plasma metabolites related to fatty acid and lipid breakdown products have been found to be elevated in mildly concussed athletes compared to controls.[27]

Other inflammatory biomarkers have yielded complex and contradictory results. However, overall some initial inflammation may promote recovery, but prolonged or high levels of inflammation could be detrimental.[28, 29]  For example, in severely brain-injured patients a group of CSF inflammatory mediators including intraleukin 6 and 8 discriminated between good versus poor 6-month outcomes with higher levels of these inflammatory mediators occuring in those with poorer outcomes.[30]

In addition to structural and chemical changes, gene expression is altered following closed head injury. Genes involving growth factors, hormones, toxin-binders, apoptosis (programmed cell death), and inflammation have all been implicated in rodent models. For example, in a mouse model of head injury, elevated levels of the transcription factor p53 were found. p53 translocates to the nucleus and initiates apoptosis or programmed cell death. Such a process could account for the delayed neuronal loss seen in head injuries.[31]  Furthermore, in humans, differential activation of inflammatory regulatory genes has been associated with the worse outcomes observed in elderly patients with closed head injuries compared to their younger counterparts.[32]  

Secondary insults

Hypotension and hypoxia cause the most prominent secondary trauma-induced brain insults. Both hypoxia and hypotension had adverse impacts on outcomes of 716 patients with severe head injuries from the Traumatic Coma Data Bank in the United States. Efforts to limit hypoxic injury with in-field intubation have been unsuccessful. Indeed, a multicenter study of 4098 patients with severe traumatic brain injury found that in-field intubation was associated with a dramatic increase in death and poor long-term neurologic outcome, even after controlling for injury severity.[33]  More current epidemiologic research supports the lack of benefit of early intubation.[34]

In the Trauma Coma Data Bank study, hypotension was even more significant than hypoxia and, by itself, was associated with a 150% increase in mortality rate. Systemic hypotension is critical because brain perfusion diminishes with lower somatic blood pressures. Brain perfusion (ie, cerebral perfusion pressure) is the difference between the mean arterial pressure and intracranial pressure. The intracranial pressure is increased in head injury by intracranial bleeding, cell death, and secondary hypoxic and ischemic injuries. Accordingly, another recent study reported that death and increased disability outcomes correlated with the durations of both systemic hypotension and elevated intracranial pressures.[35]

Severe anemia is often coexistent with head injuries, but blood transfusions have been associated with increased mortality and complications among 1250 ICU-admitted patients with brain injuries. This relationship held even after controlling for the degree of anemia.[36]  Similarly, adverse thromboembolic events occured among 200 severely head-injured patients treated wtih erythropoetin and transfusions.[37]

Finally, posttraumatic cerebral infarction occurs in up to 12% of patients with moderate and severe head injuries and is associated with a decreased Glasgow Coma Scale, low blood pressure, and herniation syndromes.[38]

Epidemiology

Frequency

United States

In the United States, 2.8 million individuals per year incur a head injury. Of these injuries, 75% are classified as mild. Between 1998 and 2000, the incidence of mild traumatic brain injury was 503 cases per 100,000 persons, with a doubling of this incidence in Native Americans and children. Between 2007 and 2012, brain injury hospitalizations, death, and emergency department visits increased from 640 to 890 cases per 100,000 persons in the United States.[1, 2]

In 2003, elderly persons with head injuries exhibited a doubling in hospitalizations and deaths compared to the national average.[39]  This trend has persisted with Canadian, European, and US data demonstrating an increased frequency and severity of traumatic brain injury in the elderly, primarily secondary to falls, while motor vehicular causes have decreased.[40, 41, 42, 2]

International

Head injury data are difficult to compare internationally for multiple reasons, including inconsistencies and complexities of diagnostic coding and inclusion criteria, case definitions, ascertainment criteria (for example, hospital admissions versus door-to-door surveys), transfers to multiple care facilities (for example, patient admissions may be counted more than once), and regional medical practices, such as the recent development in the United States of more outpatient, as opposed to inpatient, services for those with mild head injuries. Adding to this complexity is the finding that some individuals with cognitive and emotional sequelae from mild head injury may not establish the casual connection between their injury and its consequences. Such patients may not seek treatment and may not be expressed in official demographic data.[43, 44]

Despite such obstacles, a recent meta-analysis extrapolated head injury rates to total population estimates and found that Southeast Asian and Western Pacific nations carried the heaviest global head injury burden.[45]

Mortality/Morbidity

According to the CDC, about 56,000 individuals die from traumatic brain injuries each year in the United States. Almost twice that number suffer permanent disability.[2]

Race

A study of intentional head injury from Charlotte, North Carolina, found minority status was a major predictor of intentional head injury, even after controlling for other demographic factors.[46] Furthermore, worse clinical outcomes have been described for African American children with moderate-to-severe head injuries compared with their white counterparts.[47]

Sex

Men in the United States are nearly twice as likely to be hospitalized with a brain injury than women. This male predominance is found worldwide.

Age

Approximately half of the patients admitted to a hospital for head injury are aged 24 years or younger. The rates of emergency room visits for the head-injured elderly are more than 3 times higher for those over 84 years of age compared to those between 65 to 74 years of age.[48]

History

History in most patients with head injury should be self-evident. However, consider trauma with intracerebral pathology in any patient with a coma of unknown etiology.

Physical

Elemental neurologic examination

See the list below:

Bedside cognitive testing

See the list below:

Causes

Road accidents involving motor vehicle drivers and occupants, cyclists, and pedestrians are the main risk factor for head injuries. Assaults in economically depressed regions and during wartime are other major risk factors. Athletic participation, especially football and soccer, is another important cause of these injuries.

Falls cause head injuries in elderly patients and children, occasionally with catastrophic results. The incidence of fall-related traumatic brain injury has been increasing in the United States and in 2013 resulted in 12,015 deaths and 91,470 hospitalizations in the elderly.[2]

Blast injuries from incendiary devices can cause head trauma and primarily occur in soldiers, although even civilian tire explosions have been implicated.[59] While the energy from the blast can directly impact the cranium and be transferred to the brain, some researchers have hypothesized that systemic blood vessels may actually transmit the shock waves.[60] Current clinical studies, however, have failed to identify a unique pattern of neuropsychologic deficits in patients who have incurred such blast injuries.[61]

Anticoagulants and antiplatelet medications, such as aspirin, raise the risk of intracranial bleeding with even trivial head injuries.[62] For example, among elderly patients with head injuries, clopidogrel use has been associated with a 15 times greater mortality compared with patients not taking antithrombotics.[63]  Head-injured warfarin users, compared to direct oral anticoagulant users, exhibited a greater mortality and greater need for neurosurgical procedures.[64]  Curiously, another study documented counterintuitively that in patients with mild head injury, antiplatelet agent use more than doubled the risk of intracranial bleeding compared to anticoagulant use.[65]

Alcohol use raises the risks of incurring a head injury. Perhaps because it may impede excitotoxicity, alcohol use at the time of injury may actually decrease the likelihood of a poor outcome.

A newer study of intentional head injuries reported that patients consuming alcohol had higher initial GCS scores.[46] Another study of patients with apparently trivial injuries (patients either were found down or fell from heights < 10 ft) found that outcomes were better in patients who were severely intoxicated (blood alcohol levels >200 mg/dL). Methamphetamine use has also been shown to reduce mortality in severe head injury.[66] More recently, patients with severe brain injuries and high blood alcohol levels (≥ 0.08 mg/L) exhibited a significantly lesser mortality compared with patients with lower levels or the absence of alcohol in their blood.[67]

The presence of even one of the alleles for the APOE4 genotype may increase the risk of a poor outcome.

Laboratory Studies

See the list below:

Imaging Studies

CT scanning

Computerized tomography (CT) is the main imaging modality used in the acute setting.

Controversy exists as to whether all patients with mild head injuries should have neuroimaging. In general, patients with any loss of consciousness should undergo CT scanning.

Some researchers have established clinical criteria to identify those patients who are most likely to have abnormal scans. For example, in a group of 909 consecutive patients who had experienced a mild head injury with a transient loss of consciousness, yet scored a full 15 on their initial GCS, all 57 (6%) patients with abnormal CT scans were identified by the presence of any one of the following clinical features: age older than 60 years, headaches, vomiting, alcohol or drug intoxication, trauma above the clavicles, memory problems, or seizures.[86] More complicated criteria have been invoked to predict abnormal head CT scans even without loss of consciousness; however, these rules are cumbersome.[87] Further validation of such imaging rules is needed. In the specific case of the elderly with syncope and a subsequent fall, dramatically increased rates of CT abnormalities have been observed.[88]

Repeat CT is needed, of course, when clinical deterioration occurs. The need for routine repeat head CT is unclear. A 2006 multistudy review found neurosurgical interventions resulting from a repeat CT scan occurred in 0-54% of patients.[89]

In addition, emergent brain imaging may be performed for nonmedical reasons. A 10-year study of elderly women with closed head injuries revealed that in general, emergency department physicians who practice in states with tort reform laws ordered significantly less neuroimaging studies than those physicians who practice in states without such legislation.[90]

See the images below.



View Image

This 50-year-old woman with epilepsy seized and struck her head. Her initial Glasgow Coma Scale score was 12. Her scan shows prominent right temporal ....



View Image

This 40-year-old woman was anticoagulated with warfarin (Coumadin) and fell out of her hospital bed. She subsequently died. Her CT scan shows an obvio....

MRI

Magnetic resonance imaging (MRI) is typically reserved for patients who have mental status abnormalities unexplained by CT scan findings. MRI has been demonstrated to be more sensitive than CT scanning, particularly at identifying nonhemorrhagic diffuse axonal injury lesions.

MRI imaging has shown degeneration of the corpus callosum following severe head injuries with axonal damage in adults and children.[91]

Furthermore, increased total lesion volume on fluid-attenuated inversion-recovery (FLAIR) MRI images has been demonstrated to correlate with poor clinical outcomes as well.[92]

Remember that white matter hyperintensities in patients with head trauma may recede when initial MRI scans are compared with those obtained in the months following the injury.

See the image below.



View Image

This 35-year-old man was in a motor vehicle accident. His initial Glasgow Coma Scale score was 7. He had left hemiparesis. He recovered orientation to....

Diffusion tensor imaging may document axonal pathologies in patients with head injury even when conventional MRI scans are unremarkable. For example, diffusion tensor imaging has identified impaired water diffusion indicating white matter tract disruption in patients with mild head injuries whose MRI scans were normal. Cortical projection fibers were frequently abnormal, and using an innovative fiber tracking methodology, actual disruption of cortical projection fibers could be visualized in 19% of fiber groups studied.[93] Similarly, attention impairments in patients with mild head injuries have recently been correlated with diffusion abnormalities in cortical projection fibers.[94]  Furthermore, utilizing this methodology, aggresive behavior in mildly head injured patients has been correlated with reduced white matter in the corpus callosum.[95]  Finally, in severe head injuries, reduced track length and reduced track number have correlated with a worse 6-month mortality.[96]

Functional imaging and MRI spectroscopy may have eventual clinical utility. At present, they are promising research tools.

Behavioral disorders, memory, and executive dysfunction correlated with abnormalities of cingulate gyrus metabolism in 13 patients with severe head injuries who underwent resting 18F-fluorodeoxyglucose positron emission tomographic (PET) imaging and a battery of neuropsychological tests.[97] A more recent study found that while only 34% of CT results were abnormal in 92 patients with mild head injury, 63% of SPECT results demonstrated regions of hypoperfusion within 72 hours of the trauma. Frontal hypoperfusion predominated in adults.[98]

Proton magnetic resonance spectroscopy of frontal white matter that appears normal on MRI has shown a decrease in neuronal N-acetylaspartate spectra and an increase in choline spectra in patients with head injuries indicating neuronal loss.[99, 100]

Other Tests

EEG is of limited usefulness in patients with head injuries.

Histologic Findings

Although a comprehensive discussion of the histology of traumatic brain injury is beyond the scope of this article, several important newer immunohistochemical techniques have further elucidated the pathophysiology of brain injury.

Medical Care

Acute management

In the setting of acute head injury, give priority to the immediate assessment and stabilization of the airway and circulation. Despite the fact that prehospital intubation has become common, at least one study has reported a higher rate of mortality in patients intubated in the field than in those intubated in the hospital setting. In this study, however, more critically ill patients required in-field intubation.[33]

Following stabilization, direct attention to prevention of secondary injury. Keep mean arterial pressures above 90 mm Hg; arterial saturations should be greater than 90%. Urgent CT scanning is a priority.

Next, focus attention on reducing intracranial pressure, since elevated intracranial pressure is an independent predictor of poor outcome. If the intracranial pressure rises above 20-25 mm Hg, intravenous mannitol, CSF drainage, and hyperventilation can be used. Hypertonic saline has also been used in lieu of mannitol to lower intracranial pressure, but a recent meta-analysis found no evidence of diminished mortality or improved ICP control with this treatment.[110]  More definitive studies are obviously needed.[111] If the intracranial pressure does not respond to these conventional treatments, high-dose barbiturate therapy is permissible, despite the fact that no evidence currently suggests that barbiturate treatment actually improves outcomes. (Its blood pressure–lowering effects may be detrimental.)[13]

Interestingly, a 2008 study utilizing the National Trauma Data Bank retrospectively uncovered a 45% reduction in survival in patients who underwent intracranial pressure monitoring.[112] These results had been called into question because of a dearth of clinical and neuroimaging data, but a 2012 prospective study of 2134 patients with severe traumatic brain injury found improved 2-week survival in patients who underwent ICP monitoring compared to those who were not monitored. Nevertheless, the non-monitored patients may have had a more grave prognosis to start with because they were significantly older and more likely to have had pupillary abnormalities, factors which could have impacted the treating physicians' decision to implement ICP monitoring.[113]

Another approach used by some clinicians is to focus primarily on improving cerebral perfusion pressure as opposed to intracranial pressure in isolation. One study reported that 80% of patients with severe head injuries experienced recoveries with no or little disability after volume expansion, mannitol, CSF drainage, and vasopressors were used to maintain a cerebral perfusion pressure of at least 70 mm Hg.[114] Other studies have found higher perfusion pressures were associated with more complications and have recommended maintaining a cerebral perfusion pressure of 50-70 mm Hg.[115]

The question whether saline or albumin fluid resuscitation would maximize cerebral perfusion pressure and lead to improve outcomes lead to a large, double-blind, randomized controlled study of 460 patients with Glasgow Coma Scale scores < 13 who also had abnormal head CT scan results. A post-hoc 2-year follow-up demonstrated increased mortality in those receiving albumin as opposed to saline.[116]

Although hypothermic therapy initially appeared promising, and despite the fact that hypothermia decreases intracranial pressure, a large randomized study of 392 patients with head injuries recently demonstrated that hypothermic therapy does not improve outcomes. In addition, a post-hoc analysis found that the rewarming of patients with head injury who arrived in the emergency department already hypothermic was likely detrimental.[117] Furthermore, a current review of 23 randomized, controlled trials concluded that this therapy was of no benefit.[118]

Although acute hypothermic treatment has been found to worsen outcomes in patients with diffuse head injuries, it may improve outcomes in patients with surgically-evacuated hematomas. This indicates a potential benefit in this subgroup; however, further prospective studies are needed.[119]  Current opinion holds that therapeutic hypothermia administration should be reserved only for clinical trials.[120]

Head injury induces a hypermetabolic state and early nutritional interventions may be as critical as cerebral perfusion pressure. Parental or enteral feedings reduced mortality by at least 50% in one study when given early in the course of severe head injury.[121]

As mentioned previously, head injury may alter coagulation parameters, and this can raise the risk of deep venous thrombosis to as much as 15% if no pharmacologic prophylaxis is given within the first 48 hours.[122] The risk of extension of intracranial bleeding needs to be balanced with the benefits of thromboembolic prevention. A retrospective review suggested that early prophylaxis is safe because there was no difference between intracranial hemorrhage progression in patients with head injury who received enoxaparin or heparin within the first 3 days versus later in the course of their hospitalization.[123] Further studies, of course, are required.

Steroids have demonstrated no benefit in the treatment of acute head injury. A 2004 multicenter European randomized trial of steroids versus placebo found a higher mortality after only 2 weeks in the steroid-treated patients.[124]  Steroid-induced hyperglycemia may have been detrimental as a recent meta-analysis found careful glucose regulation improved functional outcomes in head-injured patients.[125]

Phenytoin has demonstrated efficacy in controlling early posttraumatic seizures, but mortality rates, surprisingly, were unaffected by this benefit. In 1 study, approximately 2.5% of patients treated with phenytoin had an allergic reaction to the drug during the first 2 weeks of therapy.[126] A trial of valproate in early seizure prophylaxis showed a trend toward an increased mortality rate. Because of its relatively benign side-effect profile, levetiracetam has been increasingly employed to prevent post-traumatic seizures, but its efficacy has not been empirically validated.[127]  Indeed, a retrospective review of 5551 acutely brain-injured patients found no significant difference in seizure rates between the patients prophylaxed with levetiracetam and the patients who were untreated, calling into question the routine practice of employing levetiracetam for seizure prophylaxis.[128] Anticonvulsant therapy, if used, should be discontinued after 1–2 weeks unless further seizures supervene.[129]

Finally, as stated previously, neuroprotective agents mostly have failed to improve the outcomes of patients with brain injury. However, the calcium channel blocker nimodipine was successful in reducing rates of death and severe disability when instituted acutely in patients with head injuries and traumatic subarachnoid hemorrhages, despite its failure to improve outcomes in 2 large trials of patients with all types of traumatic intracranial injuries.[130]

Although numerous synthetic neuroprotective agents are under development, several existing substances have shown promise, but other agents have been disappointing.

Because of its excitotoxic blocking properties, magnesium chloride has been used to reduce cortical injury in experimentally brain-injured rats. Unfortunately, a human double-blind study of 499 patients with moderate or severe head injury failed to show benefit; the magnesium-treated patients actually did worse. One potential confounder in this study was vigilance and aggressive repletion of hypomagnesemia in controls.[131]

Although promising in rodents, in a recent pair of randomized, controlled multicenter studies, the neurosteroid progesterone showed no beneficial effect on functional outcomes in patients with acute traumatic brain injury.[132, 133, 134, 135]

The first trial, the double-blind PROTECT (Progesterone for the Traumatic Brain Injury, Experimental Clinical Treatment) III study in patients with severe to moderate acute traumatic brain injury, found no significant difference in favorable outcomes between treatment and placebo groups; the trial was halted after enrollment of 882 of 1140 planned participants.[132]  Subjects in the progesterone group had higher rates of phlebitis or thrombophlebitis than those in the placebo group. In the second study, SYNAPSE (the Study of a Neuroprotective Agent, Progesterone, in Severe Traumatic Brain Injury), 1195 patients with severe traumatic brain injury were assigned to receive either progesterone or placebo.[133]  Similar rates of favorable outcomes and mortality were observed in the two groups.

Experimental brain injury creates permeability in mitochondrial membranes, which contributes to cell death by causing calcium effluxes and energy depletion. Cyclosporin inhibits mitochondrial permeability and has been used in a phase II study of patients with traumatic brain injuries. Further trials are planned.[136]

More conventional agents have also included propranolol in a large, non-randomized prospective trial. When initiated within the first 24 hours in patients with moderate to severe head injuries, propranolol significantly reduced mortality, presumable by blocking the catecholamine surge that accompanies the initial injury.[137]  A recent randomized trial of erythropoetin also showed promising effects with 33% of treated severely head-injured patients exhibiting a good recovery compared to only 13% of controls.[138]

Cannabinoids also protect against excitotoxicity, but disappointingly, in a recent phase 3 trial, dexanabinol, a weak N -methyl-D-aspartic acid (NMDA) antagonist, showed no efficacy in outcome improvement when given within 6 hours to patients with severe closed head injuries.[139] More encouraging but less rigorous was a retrospective analysis of traumatic brain-injured patients that found decreased mortality among those patients with THC in their urine compared to those without this substance.[140]

Rosuvastatin given in the acute phase of moderate head injury significantly reduced amnesia in a double-blind placebo-controlled study of 34 patients.[141]  Other research has retrospectively found that statin use was associated with a decreased mortality in a large, 100,515 patient cohort of brain injured elderly.[142]  Furthermore, a randomized prospective trial of atorvastatin demonstated improved functional outcomes at 3 months post-injury in patients with brain contusions who were treated with a mere 10-day course.[143]

Animal studies of some health food supplements may lead to new directions. The dietary supplement creatine, when fed to rats for 4 weeks prior to an experimental brain injury, reduced cortical damage by 50%, primarily through stabilizing mitochondrial functioning.[144]  Furthermore, an open-label study of children and adolescents with traumatic brain injuries reported not only a shortened duration of post-traumatic amnesia but also reduced subjective symptoms in those treated with oral creatine for 6 months compared to those not treated.[145]  Melatonin is a free-radical scavenger, and when injected early in brain-injured rats, it significantly reduced levels of lipid breakdown products.[146]

Long-term management

Hypertonicity from spasticity or dystonia with attendant muscle spasms is often disabling. Although dantrolene, baclofen, diazepam, and tizanidine are current oral medication approaches to this problem, baclofen and tizanidine are customarily preferred because of their more favorable side effect profiles.

When using these agents, careful evaluation of functional status and symptom relief is a priority since adverse effects such as sedation may be pronounced.

Intrathecal baclofen is a newer approach with reported efficacy and minimal adverse effects. One study of 17 patients with traumatic brain injuries showed improved motor tone and decreased muscle spasms with intrathecal baclofen, but whether these benefits will translate into improved functioning remains unknown.[147]

Botulinum toxin also has shown promise in decreasing hypertonia in patients with head injuries, primarily by improving passive range of motion rather than by decreasing functional disability.[148, 149]

Solid data on cognitive enhancing medications for patients with head injury are lacking. Typically, only small numbers of subjects have been used and demonstrable functional improvement has been only marginally convincing.

Despite these drawbacks, one double-blind, placebo-controlled study of methylphenidate demonstrated improved motor outcomes and attention in patients with head injuries during active treatment, but only 6 patients completed each 30-day treatment arm.[150] A 2006 double-blind, placebo-controlled study of 18 patients with closed head injuries treated with a single dose of 20 mg of methylphenidate achieved significant improvement in reaction times on a working memory test, but no other cognitive tasks significantly benefited.[151]

Donepezil treatment significantly improved visual and verbal memory as well as attentional deployment in 18 patients with head injuries of all levels of severity in a 2004 double-blind, placebo-controlled study.[152] Other less rigorous studies have also reported cognitive improvements in donepezil-treated, head-injured patients.[153]

Anecdotal reports exist of dramatic alerting responses to both levodopa and methylphenidate in patients with vegetative or comatose states. Levodopa treatment has also resulted in improvement in patients with akinesia and rigidity secondary to traumatic substantia nigral damage.[154] Furthermore, levodopa has even produced qualitative cognitive improvements in a small number of head-injured patients.[155]

Emotional lability and the pathologic laughing and crying associated with pseudobulbar palsy reportedly have responded rapidly and exquisitely to not only selective serotonin reuptake inhibitors but also possibly to dextromethorphan with quinidine.[156, 157]  Sertraline has shown efficacy in depression in mild head injury.[158] Treat other possible psychiatric complications of head injury on a patient-by-patient basis, since no extensive pharmacologic trials of this dimension of head injury have been conducted.

Nonmedical therapy

Although a full review of nonmedical therapies is beyond the scope of this article, some promising new developments have occurred in both physical and cognitive therapies.

Constraint-induced movement therapy is a form of physical therapy that emphasizes using the paralyzed arm and minimizes reliance on the unaffected extremity (patients commonly wear mittens on their unaffected arm for several hours a day). This form of treatment has resulted in significantly improved function of the paralyzed arm when used in small numbers of brain-injured patients 1-6 years after their injury.[159]

In a randomized trial in 120 military personnel with moderate-to-severe head injuries, in-hospital cognitive rehabilitation proved unsuccessful compared to a limited in-home program, but a subgroup post hoc analysis indicated that patients with unconsciousness lasting 1 hour or more had a greater functional recovery with in-hospital cognitive rehabilitation than those in the control group.[160]

Surgical Care

Traditionally, the prompt surgical evacuation of subdural hematomas in less than 4 hours was believed to be a major determinant of an optimal outcome. Indeed, a recent publication found a delay in surgery for acute subdural hematomas of over 5 hours was associated with increased mortality.[161] Nevertheless, other recent investigations have emphasized that the extent of the original intracranial injury and the generated intracranial pressures may be more important than the timing of surgery.

Consultations

In the acute setting, a consultation with a neurosurgeon is critical for patients with moderate or severe head injuries, focal neurological findings, or intracranial pathology identified on neuroimaging.

Diet

In the acute setting, nasogastric feedings may need to be initiated for patients with significant head injuries and depressed levels of consciousness or dysphagia. Careful attention to protein stores and electrolyte balance is critical during this phase of treatment.

Activity

Usually no general limitations are placed on activity. Patient-by-patient recommendations based on the individual's motoric and cognitive recovery are necessary.

Guidelines Summary

Guidelines for traumatic brain injury are undergoing constant revision with the incorporation of newly completed clinical studies and research.

The American Association of Neurological Surgeons generally does not produce specific treatment guidelines. However, the Brain Trauma Foundation recently published revised guidelines for severe traumatic brain injury, and their recommendations have been endorsed by neurosurgical professional organizations. These guidelines are based on high-to-moderate quality evidence and are summarized below:[167]

1.  If decompressive craniectomy is performed, a large frontal, temporal, and parietal one is preferred over smaller craniectomies.

2.  There is no evidence that hypothermia improves outcomes.

3.  There is insufficient evidence to support a specific hyperosmolar treatment (mannitol or hypertonic saline) for increased intracranial pressure.

4.  There is insufficient evidence to support CSF drainage.

5.  There is insufficient evidence to support prophylactic hyperventilation.

6.  There is insufficient evidence for sedatives, analgesics, or anesthetic agents.

7.  Steroids are to be avoided as they increase mortality.

8.  Enteral feeding should be initiated within the first week.

9.  Early tracheostomy placement is recommended.

10.  Povidone-iodine oral care should not be used as this may increase ARDS.

11.  There is insufficient evidence to support specific DVT-prevention strategies.

In 2013, the American Academy of Neurology[168] introduced guidelines for concussion in sports. These guidelines endorsed symptom checklists to be used by non-physician assesors to help diagnose concussions. An athlete with an identified concussion is prohibited from returning to play until the signs and symptoms of the concussion have resolved. Athletes with multiple concussions and objective neurologic or cognitive impairments are retired from play.

In 2008, the CDC recommended imaging guidelines for mild traumatic brain injury, which were re-affirmed in 2013.  A CT of the head is indicated in patients with head injury and loss of consciousness or amnesia if the patient has also had any of the following: headache, vomiting, age greater than 60 years, drug or alcohol intoxication, short-term memory loss, evidence of trauma above the clavicles, a seizure, a focal neurologic deficit, a GCS less than 15, or a coagulopathy. Such complicated decisions rules seem impractical and will hopefully be clarified with further research.[169]

Medication Summary

Medications commonly are used in the acute setting to control early seizures, reduce intracranial pressure, and correct electrolyte abnormalities. Nimodipine may be neuroprotective in the subset of patients with traumatic subarachnoid hemorrhages.

In the long-term setting, cognitive and motoric augmentation as well as the control of spasticity and emotional incontinence may require pharmacologic interventions.

Mannitol (Osmitrol, Resectisol)

Clinical Context:  May reduce subarachnoid space pressure by creating osmotic gradient between CSF in arachnoid space and plasma. Not for long-term use. Initially assess for adequate renal function in adults by administering test dose of 200 mg/kg, given IV over 3-5 min; should produce urine flow of at least 30-50 mL/h of urine over 2-3 h. Same test in children should produce urine flow of at least 1 mL/kg/h over 1-3 h.

Class Summary

These agents may help reduce intracranial pressure.

Phenytoin (Dilantin, Phenytek)

Clinical Context:  May act in motor cortex, where it may inhibit spread of seizure activity; activity of brainstem centers responsible for tonic phase of grand mal seizures also may be inhibited.

Individualize dose. Administer larger dose in evening if dose cannot be divided equally.

Class Summary

These agents may help prevent early seizures in head injury.

Magnesium sulfate

Clinical Context:  Nutritional supplement in hyperalimentation; cofactor in enzyme systems involved in neurochemical transmission and muscular excitability. In adults, 60-180 mEq of potassium, 10-30 mEq of magnesium, and 10-40 mmol of phosphate per day may be necessary for optimum metabolic response.

Class Summary

Magnesium is given in hypomagnesemic states to ensure that adequate stores are present during acute phase of head injuries.

Pentobarbital (Nembutal)

Clinical Context:  Short-acting barbiturate with sedative, hypnotic, and anticonvulsant properties. Can produce all levels of CNS depression.

Class Summary

These agents may help reduce intracranial pressure that is refractory to other conventional measures.

Nimodipine (Nymalize)

Clinical Context:  Indicated for improvement of neurological impairments resulting from spasms following subarachnoid hemorrhage caused by ruptured congenital intracranial aneurysm in patients who are in good neurological condition postictus.

While studies show benefit on severity of neurological deficits caused by cerebral vasospasm following subarachnoid hemorrhage, no evidence that drug either prevents or relieves spasms of cerebral arteries. Thus, actual mechanism of action unknown.

Therapy should start within 96 h of subarachnoid hemorrhage. If capsule cannot be swallowed because patient undergoing surgery or unconscious, a hole can be made at both ends of capsule with 18-gauge needle and contents extracted into a syringe. Contents then can be emptied into patients' in situ nasogastric tube and washed down tube with 30 mL isotonic saline.

Class Summary

Nimodipine has been demonstrated to improve outcomes of patients with traumatic subarachnoid hemorrhages.

Methylphenidate (Ritalin, Aptensio XR, Concerta, Daytrana, Methadate)

Clinical Context:  Blocks the reuptake of norepinephrine and dopamine into presynaptic neurons. May stimulate cerebral cortex and subcortical structures.

Class Summary

These agents may help increase alertness and some aspects of cognitive functioning in patients with brain injury.

Levodopa (Dopar, Larodopa)

Clinical Context:  Large neutral amino acid absorbed in proximal small intestine by saturable carrier-mediated transport system. Absorption decreased by meals, which include other large neutral amino acids. Only patients with meaningful motor fluctuations need consider low-protein or protein-redistributed diet. Greater consistency of absorption achieved when levodopa taken 1 h or more after meals. Nausea often reduced if levodopa taken immediately following meals. Some patients with nausea benefit from additional carbidopa in doses up to 200 mg/d. Half-life of levodopa/carbidopa approximately 2 h.

When more carbidopa required, substitute 1 25/100 tab for each 10/100 tab; when more levodopa required, substitute 25/250 tab for 25/100 or 10/100 tab.

Sustained release (SR) formulation of levodopa/carbidopa is absorbed more slowly and provides more sustained levodopa levels than immediate release (IR) dosage form; SR as effective as IR formulation when levodopa initially required and may be more convenient when fewer intakes are desired.

Patients with dissipating motor fluctuations and no dyskinesia often benefit from prolongation of short-duration response when switched from IR to SR; however, patients with meaningful fluctuations and dyskinesia often experience increase in dyskinesia when switched to SR formulation.

Doses and dosing intervals of SR form may be increased or decreased based on response; most patients have been treated adequately with 2-8 tab/d (divided doses) at intervals of 4-8 h while awake; higher doses (>12 tab/d) and intervals < 4 h have been used but usually are not recommended; if < 4-h interval used or if divided doses are not equal, give smaller doses at end of day. Allow at least a 3-d interval between dosage adjustments. May administer as whole or half tab, which should not be crushed or chewed.

Class Summary

These agents may increase alertness in patients with brain injury; also may help in occasional patients with posttraumatic parkinsonism.

Sertraline (Zoloft)

Clinical Context:  SSRIs are antidepressant agents that are chemically unrelated to TCAs, tetracyclic antidepressants (TeCAs), or other available antidepressants. They inhibit central nervous system (CNS) neuronal uptake of serotonin and may have a weak effect on neuronal reuptake of norepinephrine and dopamine.

Class Summary

These agents have been of benefit in patients with head injuries and emotional incontinence.

Tizanidine hydrochloride (Zanaflex)

Clinical Context:  Tizanidine is a centrally acting muscle relaxant that is metabolized in the liver and excreted in urine and feces. A single oral dose of 8mg reduces muscle tone in patients with spasticity for several hours. Blood levels and the spasmolytic effect are linearly correlated.

Baclofen (Lioresal)

Clinical Context:  May induce hyperpolarization of afferent terminals and inhibit both monosynaptic and polysynaptic reflexes at spinal level. Baclofen presynaptically inhibits the nerve terminal. It is centrally acting and can be administered intrathecally or orally. Baclofen is the preferred drug for spasticity related to spinal cord injury (SCI). Tolerance can occur. Adverse effects are minimized if the drug is given intrathecally.

Dantrolene (Dantrium)

Clinical Context:  This is a peripherally acting medication that prevents calcium release from the sarcoplasmic reticulum. It is particularly effective in cerebral-origin spasticity, such as that occurring in traumatic brain injury (TBI), stroke, or cerebral palsy. Stimulates muscle relaxation by modulating skeletal muscle contractions at site beyond myoneural junction and acting directly on muscle.

Diazepam (Valium, Diazepam Intensol)

Clinical Context:  Diazepam acts presynaptically and is a gamma-aminobutyric acid ̶ A (GABA-A) agonist. It is centrally acting. Tolerance and addiction can occur.Depresses all levels of CNS, possibly by increasing activity of GABA. Individualize dosage and increase cautiously to avoid adverse effects.

Class Summary

Antispasticity medications can be useful. These agents may reduce painful cramping and detrimental muscle tightening. However, one of the drawbacks of using these agents is that some patients find that the stiffness of spasticity helps them to overcome the muscle weakness. When patients are medicated to reduce stiffness, walking may become more difficult. Adverse effects can also be a problem.

If the patient does well with the medications, however, discomfort associated with spasticity can generally be reduced, mobility can be improved, and the effectiveness of physical therapy (PT) can be enhanced.

Dextromethorphan/quinidine (Nuedexta)

Clinical Context:  Dextromethorphan is a sigma-1 receptor agonist and an uncompetitive N-methyl-D-aspartate (NMDA) receptor antagonist. Quinidine increases plasma levels of dextromethorphan by competitively inhibiting cytochrome P4502D6, which catalyzes a major biotransformation pathway for dextromethorphan. The mechanism by which dextromethorphan exerts therapeutic effects is unknown.

Class Summary

Glutamate release inhibition and glutamate receptor blockade are alternatives to potentiating D2 receptors in the indirect pallidal outflow pathway by reducing the glutamate-related excitatory circuit in the outflow pathway.

Further Outpatient Care

This also depends on the degree of the head injury and the individual patient's cognitive and motor abilities and pain complaints.

Further Inpatient Care

This needs to be individualized. Certainly after a moderate or severe head injury, transfer to an inpatient rehabilitation unit is recommended.

Transfer

Patients with moderate or severe head injuries and head injuries with significant extracranial components are cared for best at a specialized trauma center.

Deterrence/Prevention

Reducing morbidity and mortality rates associated with head injuries is likely to be difficult. Violence, automobiles, and drug and alcohol use are prevalent.

A study of community-based programs reported modest success, primarily by employing increased police surveillance and law enforcement to reduce overdrinking and alcohol-related injuries. Motor vehicle accidents in which the driver was intoxicated declined 6%, and more significantly, overall assault cases seen in local emergency departments decreased 42%.[170]

Another study has shown that patients who were screened for alcohol problems and provided with an organized intervention to reduce their alcohol consumption exhibited a 47% decrease in emergency department–evaluated injuries compared to patients receiving no alcohol screening or intervention.[171]

The use of protective devices also is promising. A meta-analysis of case-control studies of bicycle helmet use concluded that helmets reduce the risk of severe head and brain injuries by 63–88%.[172] Indeed, a 2006 study of 160 cyclist injuries in Singapore found that helmet users sustained head injuries only 5.9% of the time, compared with 40% of the time for nonusers.[173] Similarly, a California law mandating the use of bicycle helmets for riders aged 17 years and younger reduced traumatic brain injuries by 18%. However, subgroup analysis revealed that this reduction failed to apply to urban, female, and African American riders.[174]

Falls in the elderly are often multifactorial in origin and consequently, solutions are likely to be complex. Nevertheless, random allocation of indepently living elderly patients to a fall-prevention regime of strength and balance training reduced both the number of falls as well as their severity.[175]

Complications

Mild head injury

Mild head injuries are those that generate GCS scores of 13-15. Such injuries usually are considered relatively benign, and the accompanying cognitive impairments typically resolve within 3 months of injury.

Patients with lingering complaints are often assumed to have either a psychological reaction to the injury or to be malingering. Various standardized neuropsychologic instruments are available to help sort out legitimate from illegitimate cognitive impairments. About 25% of patients with mild head injury taking such tests yield invalid profiles.[176] Furthermore, coexisting musculoskeletal injuries may impact cognitive testing. For example, collegiate athletes with musculoskeletal injuries performed just as poorly on computerized neuropsychological tests as athletes with concussions.[177]

However, an initial grading of mild does not necessarily mean a mild outcome. As many as 3% of patients with an initial mild injury may require a neurosurgical operation.[178] Some patients have died hours after sustaining trivial head injuries. Also, as previously mentioned, axonal damage has been documented pathologically with mild head injuries.

Disability rates may be pronounced with putatively mild injuries as well. Recent studies have demonstrated that following mild head injury, only 54-79% of patients are able to return to full preinjury employment. One study of 148 patients with mild head injury based on the GCS discovered that after 1 year, 26% had moderate disability and 3% had severe disability, but all these patients also had either radiological abnormalities or focal neurological signs, placing them in the more severe range of mild head injuries.[179] A recent review sponsored by the National Institute of Medicine concluded that there is no clear evidence of lasting cognitive impairments attributable to mild closed head injuries.[180]

Second impact syndrome

In the United States, athletic competitions account for 300,000 mild head injuries per year. The second impact syndrome occurs when an athlete suffers a minor concussion and subsequently is re-injured in play. The repeated concussive events are theorized to result in autoregulatory dysfunction and vascular congestion. Catastrophic brain edema, herniation, and sudden death may ensue.

At least 35 cases occurred among US football participants from 1980–1993, but the general incidence of this syndrome is unknown. Concerns about athletes at risk returning to play too soon have generated formalized recommendations from the American Academy of Neurology. Return to play is postponed for increasing lengths of time depending on the severity of the concussion.[181, 182]  In addition, return to play is contingent on the resolution of the initial concussion symptoms.[168]

Some researchers have questioned the existing literature's documentation of initial injuries, hypothesizing that the second impact syndrome is more one of primary impact and that secondary prevention strategies are not justified empirically.[183] Other researchers more recently have stressed the prolonged nature of recovery from athletic head injuries and the need for longer recuperation prior to return to play.[184]  American football, male gender, and young age ranges are associated with this syndrome.[185]

Posttraumatic epilepsy

Posttraumatic seizures occur clinically in approximately 4% of patients with head injuries within the first week of the injury. Continuous EEG monitoring may disclose a higher incidence (22%).[101, 102]

Seizures after the first week occur in 4-30% of patients. The severity of the head injury, early seizures, depressed skull fractures, and temporal and frontal injuries identified on CT scans all have been associated with the development of late seizures.[186]

Although focal EEG findings traditionally have not been predictive of late seizures, one study reported that a focal EEG 1 month after injury resulted in a 3.49-times higher risk of posttraumatic epilepsy.[187]

Recently, MRI-visualized hippocampal sclerosis has been associated with intractable epilepsy in patients who sustained moderate-to-severe head injuries when aged 10-31 years.[188]

Posttraumatic headaches

Posttraumatic headaches are common and may occur in 30-90% of patients after a head injury.[189] The alterations in cations, catecholamines, and excitatory amino acids are similar in both migraine and head injury.[190]

Posttraumatic headaches typically manifest with a vascular component, but chronic daily headaches are also common.

Although controversial, some authors have reported that most posttraumatic headaches are primarily rebound or analgesic-overuse headaches. Nearly three fourths of such patients may benefit from cessation of pain medications.[191]

Greater occipital neuralgia can occur following head and neck injuries. Greater occipital nerve pain occurs in the back of the head and may be characterized by lancinating or aching sensations in this region.

Posttraumatic movement disorders

Tremor, dystonia, parkinsonism, myoclonus, and hemiballism all can occur following head injuries.

In a 2-year follow-up study of 398 patients with severe head injuries, 12% had persistent movement disorders. Disabling dystonia and low-frequency kinetic tremors were present in 5.4%. Parkinsonism and myoclonus attributable to the injury occurred in less than 1% of patients.[53]

Posttraumatic psychiatric disorders

Disorders of emotional functioning have been documented repeatedly after head injuries. A case-control study of 91 patients hospitalized with traumatic brain injury recorded a 33% incidence of major depression.[192] Depression has been associated with left frontal injuries. Using a questionnaire methodology, 56% of 774 head injured patients of all levels of severity exhibited depression 10 weeks after their injury.[193]  Bipolar disorder is also more frequent in patients with head injuries than in the general population and is associated with seizures and right hemispheric lesions.

Additionally, impulsive and disinhibited behaviors are common in patients with frontal injuries, although even obsessive-compulsive features have also been reported.[194]

Head injury-related psychosis is controversial. A case-control study of 45 patients with psychosis following head injury found that auditory hallucinations and paranoid delusions developed after a 54-month postinjury latent period. More widespread injury on neuroimaging and decreased cognitive functioning characterized the psychotic patients with head injuries compared with nonpsychotic control patients with head injuries.[195]

Prognosis

This discussion has delineated a myriad of prognostic factors. Head injuries may result in death, a vegetative state, partial recovery, or full return to work. Each patient presents with a unique baseline neurological make up, mechanisms of injury, secondary complications, and postinjury adjustment and support system.

The most important prognostic factors are probably age, mechanism of injury, postresuscitation GCS score, postresuscitation pupillary reactivity, postresuscitation blood pressures, intracranial pressures, duration of posttraumatic amnesia or confusion, sitting balance, and intracranial pathology identified on neuroimaging.

The mortality rate of severe head injuries ranges from 25-36% in adults within the first 6 months after injury. Most deaths occur within the first 2 weeks.

Patient Education

The physician may be hesitant to suggest to patients that problems may arise from a mild head injury. Such information may induce the expectations of symptoms when no symptoms are present and arouse anxiety. However, at least one study has shown that patients with head injury who were contacted by phone and offered education about their injury and follow-up care experienced significantly fewer postconcussive symptoms and less disruption of social activities.[205]

At present, most patients incurring a head injury probably should be informed that cognitive and emotional dysfunction as well as head pain and other somatic symptoms are not uncommon in the aftermath. At least in mild injuries, these symptoms typically are self-limited, and most people return to normal functioning after a few weeks to months.

For excellent patient education resources, visit eMedicineHealth's First Aid and Injuries Center, Brain and Nervous System Center, and Eye and Vision Center. Also, see eMedicineHealth's patient education articles Concussion, Dementia in Head Injury, and Black Eye.

Author

David A Olson, MD, Clinical Neurologist, Dekalb Neurology Group, Decatur, Georgia

Disclosure: Nothing to disclose.

Specialty Editors

Francisco Talavera, PharmD, PhD, Adjunct Assistant Professor, University of Nebraska Medical Center College of Pharmacy; Editor-in-Chief, Medscape Drug Reference

Disclosure: Received salary from Medscape for employment. for: Medscape.

Florian P Thomas, MD, PhD, MA, MS, Chair, Neuroscience Institute and Department of Neurology, Director, National MS Society Multiple Sclerosis Center and Hereditary Neuropathy Foundation Center of Excellence, Hackensack University Medical Center; Founding Chair and Professor, Department of Neurology, Hackensack Meridian School of Medicine at Seton Hall University; Professor Emeritus, Department of Neurology, St Louis University School of Medicine; Editor-in-Chief, Journal of Spinal Cord Medicine

Disclosure: Nothing to disclose.

Chief Editor

Stephen A Berman, MD, PhD, MBA, Professor of Neurology, University of Central Florida College of Medicine

Disclosure: Nothing to disclose.

Additional Contributors

Joseph Carcione, Jr, DO, MBA, Consultant in Neurology and Medical Acupuncture, Medical Management and Organizational Consulting, Central Westchester Neuromuscular Care, PC; Medical Director, Oxford Health Plans

Disclosure: Nothing to disclose.

References

  1. Rates of TBI-related Emergency Department Visits, Hospitalizations, and Deaths--United States, 2001-2010. Centers for Disease Control and Prevention. Available at http://www.cdc.gov/traumaticbraininjury/data/rates.html. Accessed: September 11, 2016.
  2. Taylor CA, Bell JM, Breiding MJ, Xu L. Traumatic Brain Injury-Related Emergency Department Visits, Hospitalizations, and Deaths - United States, 2007 and 2013. MMWR Surveill Summ. 2017 Mar 17. 66 (9):1-16. [View Abstract]
  3. Wu AP, Davidson T. Posttraumatic anosmia secondary to central nervous system injury. Am J Rhinol. Nov-Dec/2008. 22:606-7.
  4. Bhatoe HS. Primary brainstem injury: benign course and improved survival. Acta Neurochir (Wien). 1999. 141(5):515-9. [View Abstract]
  5. Munjal SK, Panda NK, Pathak A. Dynamics of Hearing Status in Closed Head Injury. J Neurotrauma. Feb/2010. 27:309-316.
  6. Mackay LE, Morgan AS, Bernstein BA. Factors affecting oral feeding with severe traumatic brain injury. J Head Trauma Rehabil. 1999 Oct. 14(5):435-47. [View Abstract]
  7. Ellenberg JH, Levin HS, Saydjari C. Posttraumatic Amnesia as a predictor of outcome after severe closed head injury. Prospective assessment. Arch Neurol. 1996 Aug. 53(8):782-91. [View Abstract]
  8. Crevits L, Hanse MC, Tummers P, et al. Antisaccades and remembered saccades in mild traumatic brain injury. J Neurol. 2000 Mar. 247(3):179-82. [View Abstract]
  9. Tisdall M, Crocker M, Watkiss J, Smith M. Disturbances of sodium in critically ill adult neurologic patients: a clinical review. J Neurosurg Anesthesiol. 2006 Jan. 18(1):57-63. [View Abstract]
  10. Olivecrona M, Koskinen LD. Comment on: Early CSF and serum S 100B concentrations for outcome prediction in traumatic brain injury and subarachoid haemorrhage. Clin Neurol Neurosurg. 2016 Aug 20. [View Abstract]
  11. Herrmann M, Curio N, Jost S, et al. Release of biochemical markers of damage to neuronal and glial brain tissue is associated with short and long term neuropsychological outcome after traumatic brain injury. J Neurol Neurosurg Psychiatry. 2001 Jan. 70(1):95-100. [View Abstract]
  12. Voelker R. Taking a Closer Look at the Biomarker Test for Mild Traumatic Brain Injury. JAMA. 2018 May 22. 319 (20):2066-2067. [View Abstract]
  13. Roberts I. Barbiturates for acute traumatic brain injury. Cochrane Database Syst Rev. 2000. CD000033. [View Abstract]
  14. Stein SC. Minor head injury: 13 is an unlucky number. J Trauma. 2001 Apr. 50(4):759-60. [View Abstract]
  15. Matsuyama T, Shimomura T, Okumura Y, et al. Acute subdural hematomas due to rupture of cortical arteries: a study of the points of rupture in 19 cases. Surg Neurol. 1997 May. 47(5):423-7. [View Abstract]
  16. Eisenberg HM, Gary HE Jr, Aldrich EF, et al. Initial CT findings in 753 patients with severe head injury. A report from the NIH Traumatic Coma Data Bank. J Neurosurg. 1990 Nov. 73(5):688-98. [View Abstract]
  17. Maxwell WL, MacKinnon MA, Smith DH, et al. Thalamic nuclei after human blunt head injury. J Neuropathol Exp Neurol. 2006 May. 65(5):478-88. [View Abstract]
  18. Blumbergs PC, Scott G, Manavis J, et al. Topography of axonal injury as defined by amyloid precursor protein and the sector scoring method in mild and severe closed head injury. J Neurotrauma. 1995 Aug. 12(4):565-72. [View Abstract]
  19. Jennett B, Adams JH, Murray LS, et al. Neuropathology in vegetative and severely disabled patients after head injury. Neurology. 2001 Feb 27. 56(4):486-90. [View Abstract]
  20. Markianos M, Seretis A, Kotsou S, et al. CSF neurotransmitter metabolites and short-term outcome of patients in coma after head injury. Acta Neurol Scand. 1992 Aug. 86(2):190-3. [View Abstract]
  21. Bullock R, Zauner A, Woodward JJ, et al. Factors affecting excitatory amino acid release following severe human head injury. J Neurosurg. 1998 Oct. 89(4):507-18. [View Abstract]
  22. DeGraba TJ, Pettigrew LC. Why do neuroprotective drugs work in animals but not humans?. Neurol Clin. 2000 May. 18(2):475-93. [View Abstract]
  23. Jonas W, Lin Y, Tortella F. Neuroprotection from glutamate toxicity with ultra-low dose glutamate. Neuroreport. 2001 Feb 12,. 12(2):335-9. [View Abstract]
  24. Yang SY, Gao ZX. Determination and clinical significance of plasma levels of prostaglandins in patients with acute brain injury. Surg Neurol. 1999 Sep. 52(3):238-45. [View Abstract]
  25. Zubkov AY, Lewis AI, Raila FA, et al. Risk factors for the development of post-traumatic cerebral vasospasm. Surg Neurol. 2000 Feb. 53(2):126-30. [View Abstract]
  26. Al-Mufti F, Amuluru K, Changa A, Lander M, Patel N, Wajswol E, et al. Traumatic brain injury and intracranial hemorrhage-induced cerebral vasospasm: a systematic review. Neurosurg Focus. 2017 Nov. 43 (5):E14. [View Abstract]
  27. Fiandaca MS, Mapstone M, Mahmoodi A, Gross T, Macciardi F, Cheema AK, et al. Plasma metabolomic biomarkers accurately classify acute mild traumatic brain injury from controls. PLoS One. 2018. 13 (4):e0195318. [View Abstract]
  28. Kumar RG, Diamond ML, Boles JA, Berger RP, Tisherman SA, Kochanek PM, et al. Acute CSF interleukin-6 trajectories after TBI: associations with neuroinflammation, polytrauma, and outcome. Brain Behav Immun. 2015 Mar. 45:253-62. [View Abstract]
  29. Russo MV, McGavern DB. Inflammatory neuroprotection following traumatic brain injury. Science. 2016 Aug 19. 353 (6301):783-5. [View Abstract]
  30. Nwachuku EL, Puccio AM, Adeboye A, Chang YF, Kim J, Okonkwo DO. Time course of cerebrospinal fluid inflammatory biomarkers and relationship to 6-month neurologic outcome in adult severe traumatic brain injury. Clin Neurol Neurosurg. 2016 Oct. 149:1-5. [View Abstract]
  31. Plesnila N, von Baumgarten L, Retiounskaia M, Engel D, Ardeshiri A, Zimmermann R, et al. Delayed neuronal death after brain trauma involves p53-dependent inhibition of NF-kappaB transcriptional activity. Cell Death Differ. 2007 Aug. 14(8):1529-41. [View Abstract]
  32. Cho YE, Latour LL, Kim H, Turtzo LC, Olivera A, Livingston WS, et al. Older Age Results in Differential Gene Expression after Mild Traumatic Brain Injury and Is Linked to Imaging Differences at Acute Follow-up. Front Aging Neurosci. 2016. 8:168. [View Abstract]
  33. Wang HE, Peitzman AB, Cassidy LD, et al. Out-of-hospital endotracheal intubation and outcome after traumatic brain injury. Ann Emerg Med. 2004 Nov. 44(5):439-50. [View Abstract]
  34. Haltmeier T, Schnüriger B, Benjamin E, Brodmann Maeder M, Künzler M, Siboni S, et al. Isolated blunt severe traumatic brain injury in Bern, Switzerland, and the United States: A matched cohort study. J Trauma Acute Care Surg. 2016 Feb. 80 (2):296-301. [View Abstract]
  35. Lannoo E, Van Rietvelde F, Colardyn F, et al. Early predictors of mortality and morbidity after severe closed head injury. J Neurotrauma. 2000 May. 17(5):403-14. [View Abstract]
  36. Salim A, Hadjizacharia P, DuBose J, Brown C, Inaba K, Chan L. Role of anemia in traumatic brain injury. J Am Coll Surg. 2008 Sep. 207(3):398-406. [View Abstract]
  37. Robertson CS, Hannay HJ, Yamal JM, et al. Effect of erythropoietin and transfusion threshold on neurological recovery after traumatic brain injury: a randomized clinical trial. JAMA. 2014 Jul 2. 312 (1):36-47. [View Abstract]
  38. Tian HL, Geng Z, Cui YH, Hu J, Xu T, Cao HL. Risk factors for posttraumatic cerebral infarction in patients with moderate or severe head trauma. Neurosurg Rev. 2008 Oct. 31(4):431-6; discussion 436-7. [View Abstract]
  39. Rutland-Brown W, Langlois JA, Thomas KE, Xi YL. Incidence of traumatic brain injury in the United States, 2003. J Head Trauma Rehabil. 2006 Nov-Dec. 21(6):544-8. [View Abstract]
  40. Büchele G, Rapp K, König HH, Jaensch A, Rothenbacher D, Becker C, et al. The Risk of Hospital Admission Due to Traumatic Brain Injury Is Increased in Older Persons With Severe Functional Limitations. J Am Med Dir Assoc. 2016 Jul 1. 17 (7):609-12. [View Abstract]
  41. Fu TS, Jing R, McFaull SR, Cusimano MD. Recent trends in hospitalization and in-hospital mortality associated with traumatic brain injury in Canada: A nationwide, population-based study. J Trauma Acute Care Surg. 2015 Sep. 79 (3):449-54. [View Abstract]
  42. Peeters W, van den Brande R, Polinder S, Brazinova A, Steyerberg EW, Lingsma HF, et al. Epidemiology of traumatic brain injury in Europe. Acta Neurochir (Wien). 2015 Oct. 157 (10):1683-96. [View Abstract]
  43. Thurman D, Guerrero J. Trends in hospitalization associated with traumatic brain injury. JAMA. 1999 Sep 8. 282(10):954-7. [View Abstract]
  44. Cassidy JD, Carroll LJ, Peloso PM, Borg J, von Holst H, Holm L. Incidence, risk factors and prevention of mild traumatic brain injury: results of the WHO Collaborating Centre Task Force on Mild Traumatic Brain Injury. J Rehabil Med. 2004 Feb. (43 Suppl):28-60. [View Abstract]
  45. Dewan MC, Rattani A, Gupta S, Baticulon RE, Hung YC, Punchak M, et al. Estimating the global incidence of traumatic brain injury. J Neurosurg. 2018 Apr 27. 1-18. [View Abstract]
  46. Wagner AK, Sasser HC, Hammond FM, et al. Intentional traumatic brain injury: epidemiology, risk factors, and associations with injury severity and mortality. J Trauma. 2000 Sep. 49(3):404-10. [View Abstract]
  47. Haider AH, Efron DT, Haut ER, DiRusso SM, Sullivan T, Cornwell EE 3rd. Black children experience worse clinical and functional outcomes after traumatic brain injury: an analysis of the National Pediatric Trauma Registry. J Trauma. 2007 May. 62(5):1259-62; discussion 1262-3. [View Abstract]
  48. Albrecht JS, Hirshon JM, McCunn M, Bechtold KT, Rao V, Simoni-Wastila L, et al. Increased Rates of Mild Traumatic Brain Injury Among Older Adults in US Emergency Departments, 2009-2010. J Head Trauma Rehabil. 2016 Sep-Oct. 31 (5):E1-7. [View Abstract]
  49. Lau BC, Kontos AP, Collins MW, Mucha A, Lovell MR. Which On-field Signs/Symptoms Predict Protracted Recovery From Sport-Related Concussion Among High School Football Players?. Am J Sports Med. 2011 Nov. 39(11):2311-8. [View Abstract]
  50. Bernal-Sprekelsen M, Bleda-Vazquez C, Carrau RL. Ascending meningitis secondary to traumatic cerebrospinal fluid leaks. Am J Rhinol. 2000 Jul-Aug. 14(4):257-9. [View Abstract]
  51. Tien HC, Cunha JR, Wu SN, et al. Do trauma patients with a Glasgow Coma Scale score of 3 and bilateral fixed and dilated pupils have any chance of survival?. J Trauma. 2006 Feb. 60(2):274-8. [View Abstract]
  52. Mauritz W, Leitgeb J, Wilbacher I, et al. Outcome of brain trauma patients who have a Glasgow Coma Scale score of 3 and bilateral fixed and dilated pupils in the field. European Journal of Emergency Medicine. 2009. 16:153-158.
  53. Krauss JK, Trankle R, Kopp KH. Post-traumatic movement disorders in survivors of severe head injury. Neurology. 1996 Dec. 47(6):1488-92. [View Abstract]
  54. Matser JT, Kessels AG, Jordan BD, et al. Chronic traumatic brain injury in professional soccer players. Neurology. 1998 Sep. 51(3):791-6. [View Abstract]
  55. Lobato RD, Rivas JJ, Gomez PA, et al. Head-injured patients who talk and deteriorate into coma. Analysis of 211 cases studied with computerized tomography. J Neurosurg. 1991 Aug. 75(2):256-61. [View Abstract]
  56. Stuss DT, Binns MA, Carruth FG, et al. The acute period of recovery from traumatic brain injury: posttraumatic amnesia or posttraumatic confusional state?. J Neurosurg. 1999 Apr. 90(4):635-43. [View Abstract]
  57. Leininger BE, Gramling SE, Farrell AD, et al. Neuropsychological deficits in symptomatic minor head injury patients after concussion and mild concussion. J Neurol Neurosurg Psychiatry. 1990 Apr. 53(4):293-6. [View Abstract]
  58. Ruffolo LF, Guilmette TJ, Willis GW. Comparison of time and error rates on the trail making test among patients with head injuries, experimental malingerers, patients with suspect effort on testing, and normal controls. Clin Neuropsychol. 2000 May. 14(2):223-30. [View Abstract]
  59. Hefny AF, Eid HO, Abu-Zidan FM. Severe tyre blast injuries during servicing. Injury. 2009 May. 40(5):484-7. [View Abstract]
  60. Bhattacharjee Y. Neuroscience. Shell shock revisited: solving the puzzle of blast trauma. Science. 2008 Jan 25. 319(5862):406-8. [View Abstract]
  61. Belanger HG, Kretzmer T, Yoash-Gantz R, Pickett T, Tupler LA. Cognitive sequelae of blast-related versus other mechanisms of brain trauma. J Int Neuropsychol Soc. 2009 Jan. 15(1):1-8. [View Abstract]
  62. Reymond MA, Marbet G, Radu EW, et al. Aspirin as a risk factor for hemorrhage in patients with head injuries. Neurosurg Rev. 1992. 15(1):21-5. [View Abstract]
  63. Wong DK, Lurie F, Wong LL. The effects of clopidogrel on elderly traumatic brain injured patients. J Trauma. Dec/2008. 65:1303-8.
  64. Feeney JM, Santone E, DiFiori M, Kis L, Jayaraman V, Montgomery SC. Compared to warfarin, direct oral anticoagulants are associated with lower mortality in patients with blunt traumatic intracranial hemorrhage: A TQIP study. J Trauma Acute Care Surg. 2016 Sep 3. [View Abstract]
  65. Uccella L, Zoia C, Bongetta D, Gaetani P, Martig F, Candrian C, et al. Are Antiplatelet and Anticoagulants Drugs A Risk Factor for Bleeding in Mild Traumatic Brain Injury?. World Neurosurg. 2018 Feb. 110:e339-e345. [View Abstract]
  66. O'Phelan K, McArthur DL, Chang CW, Green D, Hovda DA. The impact of substance abuse on mortality in patients with severe traumatic brain injury. J Trauma. 2008 Sep. 65(3):674-7. [View Abstract]
  67. Talving P, Plurad D, Barmparas G, et al. Isolated severe traumatic brain injuries: association of blood alcohol levels with the severity of injuries and outcomes. J Trauma. Feb/2010. 68:357-62.
  68. Friedman G, Froom P, Sazbon L, et al. Apolipoprotein E-epsilon4 genotype predicts a poor outcome in survivors of traumatic brain injury. Neurology. 1999 Jan 15. 52(2):244-8. [View Abstract]
  69. Kutner KC, Erlanger DM, Tsai J, et al. Lower cognitive performance of older football players possessing apolipoprotein E epsilon4. Neurosurgery. 2000 Sep. 47(3):651-7; discussion 657-8. [View Abstract]
  70. Yue JK, Robinson CK, Burke JF, et al. Apolipoprotein E epsilon 4 (APOE-ε4) genotype is associated with decreased 6-month verbal memory performance after mild traumatic brain injury. Brain Behav. 2017 Sep. 7 (9):e00791. [View Abstract]
  71. Zhou W, Xu D, Peng X, Zhang Q, Jia J, Crutcher KA. Meta-analysis of APOE4 allele and outcome after traumatic brain injury. J Neurotrauma. 2008 Apr. 25(4):279-90. [View Abstract]
  72. Li L, Bao Y, He S, Wang G, Guan Y, Ma D, et al. The Association Between Apolipoprotein E and Functional Outcome After Traumatic Brain Injury: A Meta-Analysis. Medicine (Baltimore). 2015 Nov. 94 (46):e2028. [View Abstract]
  73. Jordan BD. Genetic influences on outcome following traumatic brain injury. Neurochem Res. 2007 Apr-May. 32(4-5):905-15. [View Abstract]
  74. Shee K, Lucas A, Flashman LA, Nho K, Tsongalis GJ, McDonald BC, et al. Alpha-synuclein (SNCA) polymorphisms exert protective effects on memory after mild traumatic brain injury. Neurosci Lett. 2016 Sep 6. 630:241-6. [View Abstract]
  75. Terrell TR, Abramson R, Barth JT, Bennett E, Cantu RC, Sloane R, et al. Genetic polymorphisms associated with the risk of concussion in 1056 college athletes: a multicentre prospective cohort study. Br J Sports Med. 2018 Feb. 52 (3):192-198. [View Abstract]
  76. Bacic A, Gluncic I, Gluncic V. Disturbances in plasma sodium in patients with war head injuries. Mil Med. 1999 Mar. 164(3):214-7. [View Abstract]
  77. Leonard J, Garrett RE, Salottolo K, Slone DS, Mains CW, Carrick MM, et al. Cerebral salt wasting after traumatic brain injury: a review of the literature. Scand J Trauma Resusc Emerg Med. 2015 Nov 11. 23:98. [View Abstract]
  78. Bareyre FM, Saatman KE, Raghupathi R, McIntosh TK. Postinjury treatment with magnesium chloride attenuates cortical damage after traumatic brain injury in rats. J Neurotrauma. 2000 Nov. 17(11):1029-39. [View Abstract]
  79. Halpern CH, Reilly PM, Turtz AR, Stein SC. Traumatic coagulopathy: the effect of brain injury. J Neurotrauma. 2008 Aug. 25(8):997-1001. [View Abstract]
  80. Stalnacke BM, Tegner Y, Sojka P. Playing soccer increases serum concentrations of the biochemical markers of brain damage S-100B and neuron-specific enolase in elite players: a pilot study. Brain Inj. 2004 Sep. 18(9):899-909. [View Abstract]
  81. Straume-Naesheim TM, Andersen TE, Jochum M, et al. Minor head trauma in soccer and serum levels of S100B. Neurosurgery. Jun/2008. 62:1297-305.
  82. Mercier E, Tardif PA, Cameron PA, Émond M, Moore L, Mitra B, et al. Prognostic value of neuron-specific enolase (NSE) for prediction of post-concussion symptoms following a mild traumatic brain injury: a systematic review. Brain Inj. 2018. 32 (1):29-40. [View Abstract]
  83. Shahim P, Tegner Y, Marklund N, Blennow K, Zetterberg H. Neurofilament light and tau as blood biomarkers for sports-related concussion. Neurology. 2018 May 15. 90 (20):e1780-e1788. [View Abstract]
  84. Metting Z, Wilczak N, Rodiger LA, Schaaf JM, van der Naalt J. GFAP and S100B in the acute phase of mild traumatic brain injury. Neurology. 2012 May 1. 78(18):1428-33. [View Abstract]
  85. Lagerstedt L, Egea-Guerrero JJ, Bustamante A, Rodríguez-Rodríguez A, El Rahal A, Quintana-Diaz M, et al. Combining H-FABP and GFAP increases the capacity to differentiate between CT-positive and CT-negative patients with mild traumatic brain injury. PLoS One. 2018. 13 (7):e0200394. [View Abstract]
  86. Haydel MJ, Preston CA, Mills TJ, et al. Indications for computed tomography in patients with minor head injury. N Engl J Med. 2000 Jul 13. 343(2):100-5. [View Abstract]
  87. Smits M, Dippel DW, Steyerberg EW, de Haan GG, Dekker HM, Vos PE. Predicting intracranial traumatic findings on computed tomography in patients with minor head injury: the CHIP prediction rule. Ann Intern Med. 2007 Mar 20. 146(6):397-405. [View Abstract]
  88. Timler D, Dworzyński MJ, Szarpak Ł, Gaszyńska E, Dudek K, Gałązkowski R. Head Trauma in Elderly Patients: Mechanisms of Injuries and CT Findings. Adv Clin Exp Med. 2015 Nov-Dec. 24 (6):1045-50. [View Abstract]
  89. Wang MC, Linnau KF, Tirschwell DL, Hollingworth W. Utility of repeat head computed tomography after blunt head trauma: a systematic review. J Trauma. 2006 Jul. 61(1):226-33. [View Abstract]
  90. Smith-Bindman R, McCulloch CE, Ding A, Quale C, Chu PW. Diagnostic imaging rates for head injury in the ED and states' medical malpractice tort reforms. Am J Emerg Med. 2011 Jul. 29(6):656-64. [View Abstract]
  91. Levin HS, Williams DH, Valastro M, et al. Corpus callosal atrophy following closed head injury: detection with magnetic resonance imaging. J Neurosurg. 1990 Jul. 73(1):77-81. [View Abstract]
  92. Pierallini A, Pantano P, Fantozzi LM, et al. Correlation between MRI findings and long-term outcome in patients with severe brain trauma. Neuroradiology. 2000 Dec. 42(12):860-7. [View Abstract]
  93. Rutgers DR, Toulgoat F, Cazejust J, Fillard P, Lasjaunias P, Ducreux D. White matter abnormalities in mild traumatic brain injury: a diffusion tensor imaging study. AJNR Am J Neuroradiol. 2008 Mar. 29(3):514-9. [View Abstract]
  94. Niogi SN, Mukherjee P, Ghajar J, Johnson C, Kolster RA, Sarkar R. Extent of microstructural white matter injury in postconcussive syndrome correlates with impaired cognitive reaction time: a 3T diffusion tensor imaging study of mild traumatic brain injury. AJNR Am J Neuroradiol. 2008 May. 29(5):967-73. [View Abstract]
  95. Dailey NS, Smith R, Bajaj S, Alkozei A, Gottschlich MK, Raikes AC, et al. Elevated Aggression and Reduced White Matter Integrity in Mild Traumatic Brain Injury: A DTI Study. Front Behav Neurosci. 2018. 12:118. [View Abstract]
  96. Sener S, Van Hecke W, Feyen BF, Van der Steen G, Pullens P, Van de Hauwe L, et al. Diffusion Tensor Imaging: A Possible Biomarker in Severe Traumatic Brain Injury and Aneurysmal Subarachnoid Hemorrhage?. Neurosurgery. 2016 Jun 27. [View Abstract]
  97. Fontaine A, Azouvi P, Remy P, et al. Functional anatomy of neuropsychological deficits after severe traumatic brain injury. Neurology. 1999 Dec 10. 53(9):1963-8. [View Abstract]
  98. Gowda NK, Agrawal D, Bal C, et al. Technetium Tc-99m ethyl cysteinate dimer brain single-photon emission CT in mild traumatic brain injury: a prospective study. AJNR Am J Neuroradiol. 2006 Feb. 27(2):447-51. [View Abstract]
  99. Garnett MR, Blamire AM, Rajagopalan B, et al. Evidence for cellular damage in normal-appearing white matter correlates with injury severity in patients following traumatic brain injury: A magnetic resonance spectroscopy study. Brain. 2000 Jul. 123 ( Pt 7):1403-9. [View Abstract]
  100. Croall I, Smith FE, Blamire AM. Magnetic Resonance Spectroscopy for Traumatic Brain Injury. Top Magn Reson Imaging. 2015 Oct. 24 (5):267-74. [View Abstract]
  101. Vespa PM, Nuwer MR, Nenov V, et al. Increased incidence and impact of nonconvulsive and convulsive seizures after traumatic brain injury as detected by continuous electroencephalographic monitoring. J Neurosurg. 1999 Nov. 91(5):750-60. [View Abstract]
  102. Aquino L, Kang CY, Harada MY, Ko A, Do-Nguyen A, Ley EJ, et al. Is Routine Continuous EEG for Traumatic Brain Injury Beneficial?. Am Surg. 2017 Dec 1. 83 (12):1433-1437. [View Abstract]
  103. Steinbaugh LA, Lindsell CJ, Shutter LA, Szaflarski JP. Initial EEG predicts outcomes in a trial of levetiracetam vs. fosphenytoin for seizure prevention. Epilepsy Behav. 2012 Mar. 23(3):280-4. [View Abstract]
  104. Carter BG, Butt W. Review of the use of somatosensory evoked potentials in the prediction of outcome after severe brain injury. Crit Care Med. 2001 Jan. 29(1):178-86. [View Abstract]
  105. Hortobágyi T, Wise S, Hunt N, Cary N, Djurovic V, Fegan-Earl A. Traumatic axonal damage in the brain can be detected using beta-APP immunohistochemistry within 35 min after head injury to human adults. Neuropathol Appl Neurobiol. 2007 Apr. 33(2):226-37. [View Abstract]
  106. Dressler J, Hanisch U, Kuhlisch E, et al. Neuronal and glial apoptosis in human traumatic brain injury. Int J Legal Med. 2007. 121:365-375.
  107. McKee AC, Cantu RC, Nowinski CJ, et al. Chronic traumatic encephalopathy in athletes: progressive tauopathy after repetitive head injury. J Neuropathol Exp Neurol. Jul/2009. 68:709-35.
  108. Stein TD, Alvarez VE, McKee AC. Chronic traumatic encephalopathy: a spectrum of neuropathological changes following repetitive brain trauma in athletes and military personnel. Alzheimers Res Ther. 2014. 6 (1):4. [View Abstract]
  109. Shahim P, Linemann T, Inekci D, Karsdal MA, Blennow K, Tegner Y, et al. Serum Tau Fragments Predict Return to Play in Concussed Professional Ice Hockey Players. J Neurotrauma. 2016 Nov 15. 33 (22):1995-1999. [View Abstract]
  110. Berger-Pelleiter E, Émond M, Lauzier F, Shields JF, Turgeon AF. Hypertonic saline in severe traumatic brain injury: a systematic review and meta-analysis of randomized controlled trials. CJEM. 2016 Mar. 18 (2):112-20. [View Abstract]
  111. White H, Cook D, Venkatesh B. The use of hypertonic saline for treating intracranial hypertension after traumatic brain injury. Anesth Analg. 2006 Jun. 102(6):1836-46. [View Abstract]
  112. Shafi S, Diaz-Arrastia R, Madden C, Gentilello L. Intracranial pressure monitoring in brain-injured patients is associated with worsening of survival. J Trauma. 2008 Feb. 64(2):335-40. [View Abstract]
  113. Farahvar A, Gerber LM, Chiu YL, Carney N, Härtl R, Ghajar J. Increased mortality in patients with severe traumatic brain injury treated without intracranial pressure monitoring. J Neurosurg. 2012 Aug 17. [View Abstract]
  114. Rosner MJ, Rosner SD, Johnson AH. Cerebral perfusion pressure: management protocol and clinical results. J Neurosurg. 1995 Dec. 83(6):949-62. [View Abstract]
  115. White H, Venkatesh B. Cerebral perfusion pressure in neurotrauma: a review. Anesth Analg. Sep/2008. 107:979-88.
  116. The SAFE Study Investigators. Saline or Albumin for Fluid Resuscitation in Patients with Traumatic Brain Injury. NEJM. Aug/2007. 357:874-84.
  117. Clifton GL, Miller ER, Choi SC, et al. Lack of effect of induction of hypothermia after acute brain injury. N Engl J Med. 2001 Feb 22. 344(8):556-63. [View Abstract]
  118. Sydenham E, Roberts I, Alderson P. Hypothermia for traumatic head injury. Cochrane Database Syst Rev. Apr/2009. 15:CD001048.
  119. Clifton GL, Valadka A, Zygun D, et al. Very early hypothermia induction in patients with severe brain injury (the National Acute Brain Injury Study: Hypothermia II): a randomised trial. Lancet Neurol. 2011 Feb. 10(2):131-9. [View Abstract]
  120. Lazaridis C, Robertson CS. Hypothermia for Increased Intracranial Pressure: Is It Dead?. Curr Neurol Neurosci Rep. 2016 Sep. 16 (9):78. [View Abstract]
  121. Härtl R, Gerber LM, Ni Q, Ghajar J. Effect of early nutrition on deaths due to severe traumatic brain injury. J Neurosurg. 2008 Jul. 109(1):50-6. [View Abstract]
  122. Reiff DA, Haricharan RN, Bullington NM, et al. Traumatic brain injury is associated with the development of deep vein thrombosis independent of pharmacologic prophylaxis. J Trauma. May/2009. 66:1436-40.
  123. Depew AJ, Hu CK, Nguyen AC, et al. Thromboembolic prophylaxis in blunt traumatic intracranial hemorrhage: a retrospective review. Am Surg. OCt/2008. 74:906-11.
  124. Roberts I, Yates D, Sandercock P, et al. 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 Oct 9. 364(9442):1321-8. [View Abstract]
  125. Zhu C, Chen J, Pan J, Qiu Z, Xu T. Therapeutic effect of intensive glycemic control therapy in patients with traumatic brain injury: A systematic review and meta-analysis of randomized controlled trials. Medicine (Baltimore). 2018 Jul. 97 (30):e11671. [View Abstract]
  126. Temkin NR, Dikmen SS, Wilensky AJ, et al. A randomized, double-blind study of phenytoin for the prevention of post-traumatic seizures. N Engl J Med. 1990 Aug 23. 323(8):497-502. [View Abstract]
  127. Kruer RM, Harris LH, Goodwin H, Kornbluth J, Thomas KP, Slater LA, et al. Changing trends in the use of seizure prophylaxis after traumatic brain injury: a shift from phenytoin to levetiracetam. J Crit Care. 2013 Oct. 28 (5):883.e9-13. [View Abstract]
  128. Zangbar B, Khalil M, Gruessner A, Joseph B, Friese R, Kulvatunyou N, et al. Levetiracetam Prophylaxis for Post-traumatic Brain Injury Seizures is Ineffective: A Propensity Score Analysis. World J Surg. 2016 Nov. 40 (11):2667-2672. [View Abstract]
  129. Temkin NR, Dikmen SS, Anderson GD, et al. Valproate therapy for prevention of posttraumatic seizures: a randomized trial. J Neurosurg. 1999 Oct. 91(4):593-600. [View Abstract]
  130. Murray GD, Teasdale GM, Schmitz H. Nimodipine in traumatic subarachnoid haemorrhage: a re-analysis of the HIT I and HIT II trials. Acta Neurochir (Wien). 1996. 138(10):1163-7. [View Abstract]
  131. Temkin NR, Anderson GD, Winn HR, Ellenbogen RG, Britz GW, Schuster J. Magnesium sulfate for neuroprotection after traumatic brain injury: a randomised controlled trial. Lancet Neurol. 2007 Jan. 6(1):29-38. [View Abstract]
  132. Wright DW, Yeatts SD, Silbergleit R, Palesch YY, Hertzberg VS, Frankel M, et al. Very Early Administration of Progesterone for Acute Traumatic Brain Injury. N Engl J Med. 2014 Dec 10. [View Abstract]
  133. Skolnick BE, Maas AI, Narayan RK, van der Hoop RG, MacAllister T, Ward JD, et al. A Clinical Trial of Progesterone for Severe Traumatic Brain Injury. N Engl J Med. 2014 Dec 10. [View Abstract]
  134. Schwamm LH. Progesterone for Traumatic Brain Injury - Resisting the Sirens' Song. N Engl J Med. 2014 Dec 10. [View Abstract]
  135. Melville, N. A. Progesterone Fails in Traumatic Brain Injury. Medscape Medical News. Available at http://www.medscape.com/viewarticle/836443. December 11, 2014; Accessed: December 11, 2014.
  136. Empey PE, McNamara PJ, Young B, et al. Cyclosporin A disposition following acute traumatic brain injury. J Neurotrauma. 2006 Jan. 23(1):109-16. [View Abstract]
  137. Ko A, Harada MY, Barmparas G, Thomsen GM, Alban RF, Bloom MB, et al. Early propranolol after traumatic brain injury is associated with lower mortality. J Trauma Acute Care Surg. 2016 Apr. 80 (4):637-42. [View Abstract]
  138. Li ZM, Xiao YL, Zhu JX, Geng FY, Guo CJ, Chong ZL, et al. Recombinant human erythropoietin improves functional recovery in patients with severe traumatic brain injury: A randomized, double blind and controlled clinical trial. Clin Neurol Neurosurg. 2016 Sep 3. 150:80-83. [View Abstract]
  139. Maas AI, Murray G, Henney H, et al. Efficacy and safety of dexanabinol in severe traumatic brain injury: results of a phase III randomised, placebo-controlled, clinical trial. Lancet Neurol. 2006 Jan. 5(1):38-45. [View Abstract]
  140. Nguyen BM, Kim D, Bricker S, Bongard F, Neville A, Putnam B, et al. Effect of marijuana use on outcomes in traumatic brain injury. Am Surg. 2014 Oct. 80 (10):979-83. [View Abstract]
  141. Tapia-Perez JH, Sanchez-Aguilar M, Torres-Corzo JG, Gordillo-Moscoso A, Martinez-Perez P, Madeville P. Effect of rosuvastatin on amnesia and disorientation after traumatic brain injury (NCT003229758). J Neurotrauma. 2008 Aug. 25(8):1011-7. [View Abstract]
  142. Khokhar B, Simoni-Wastila L, Slejko JF, Perfetto E, Zhan M, Smith GS. Mortality and Associated Morbidities Following Traumatic Brain Injury in Older Medicare Statin Users. J Head Trauma Rehabil. 2018 Jan 30. [View Abstract]
  143. Farzanegan GR, Derakhshan N, Khalili H, Ghaffarpasand F, Paydar S. Effects of atorvastatin on brain contusion volume and functional outcome of patients with moderate and severe traumatic brain injury; a randomized double-blind placebo-controlled clinical trial. J Clin Neurosci. 2017 Oct. 44:143-147. [View Abstract]
  144. Sullivan PG, Geiger JD, Mattson MP, et al. Dietary supplement creatine protects against traumatic brain injury. Ann Neurol. 2000 Nov. 48(5):723-9. [View Abstract]
  145. Sakellaris G, Nasis G, Kotsiou M, Tamiolaki M, Charissis G, Evangeliou A. Prevention of traumatic headache, dizziness and fatigue with creatine administration. A pilot study. Acta Paediatr. 2008 Jan. 97 (1):31-4. [View Abstract]
  146. Cirak B, Rousan N, Kocak A, et al. Melatonin as a free radical scavenger in experimental head trauma. Pediatr Neurosurg. 1999 Dec. 31(6):298-301. [View Abstract]
  147. Meythaler JM, Guin-Renfroe S, Grabb P, Hadley MN. Long-term continuously infused intrathecal baclofen for spastic-dystonic hypertonia in traumatic brain injury: 1-year experience. Arch Phys Med Rehabil. 1999 Jan. 80(1):13-9. [View Abstract]
  148. Richardson D, Sheean G, Werring D, et al. Evaluating the role of botulinum toxin in the management of focal hypertonia in adults. J Neurol Neurosurg Psychiatry. 2000 Oct. 69(4):499-506. [View Abstract]
  149. Gracies JM, Brashear A, Jech R, McAllister P, Banach M, Walker H, 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 Oct. 14 (10):992-1001. [View Abstract]
  150. Plenger PM, Dixon CE, Castillo RM, et al. Subacute methylphenidate treatment for moderate to moderately severe traumatic brain injury: a preliminary double-blind placebo-controlled study. Arch Phys Med Rehabil. 1996 Jun. 77(6):536-40. [View Abstract]
  151. Kim YH, Ko MH, Na SY, et al. Effects of single-dose methylphenidate on cognitive performance in patients with traumatic brain injury: a double-blind placebo-controlled study. Clin Rehabil. 2006 Jan. 20(1):24-30. [View Abstract]
  152. Zhang L, Plotkin RC, Wang G, et al. Cholinergic augmentation with donepezil enhances recovery in short-term memory and sustained attention after traumatic brain injury. Arch Phys Med Rehabil. 2004 Jul. 85(7):1050-5. [View Abstract]
  153. Khateb A, Ammann J, Annoni JM, Diserens K. Cognition-enhancing effects of donepezil in traumatic brain injury. Eur Neurol. 2005. 54(1):39-45. [View Abstract]
  154. Bhatt M, Desai J, Mankodi A, Elias M, Wadia N. Posttraumatic akinetic-rigid syndrome resembling Parkinson's disease: a report on three patients. Mov Disord. 2000 Mar. 15(2):313-7. [View Abstract]
  155. Karli DC, Burke DT, Kim HJ, Calvanio R, Fitzpatrick M, Temple D. Effects of dopaminergic combination therapy for frontal lobe dysfunction in traumatic brain injury rehabilitation. Brain Inj. 1999 Jan. 13(1):63-8. [View Abstract]
  156. Nahas Z, Arlinghaus KA, Kotrla KJ, et al. Rapid response of emotional incontinence to selective serotonin reuptake inhibitors. J Neuropsychiatry Clin Neurosci. 1998 Fall. 10(4):453-5. [View Abstract]
  157. Garcia-Baran D, Johnson TM, Wagner J, Shen J, Geers M. Therapeutic Approach of a High Functioning Individual With Traumatic Brain Injury and Subsequent Emotional Volatility With Features of Pathological Laughter and Crying With Dextromethorphan/Quinidine. Medicine (Baltimore). 2016 Mar. 95 (12):e2886. [View Abstract]
  158. Fann JR, Uomoto JM, Katon WJ. Sertraline in the treatment of major depression following mild traumatic brain injury. J Neuropsychiatry Clin Neurosci. 2000 Spring. 12(2):226-32. [View Abstract]
  159. Page S, Levine Peter. Forced use after TBI: promoting plasticity and function through practice. Brain Inj. 2003 Aug. 17(8):675-84. [View Abstract]
  160. Salazar AM, Warden DL, Schwab K, et al. Cognitive rehabilitation for traumatic brain injury: A randomized trial. Defense and Veterans Head Injury Program (DVHIP) Study Group. JAMA. 2000 Jun 21. 283(23):3075-81. [View Abstract]
  161. Zhao H, Bai XJ. Influence of operative timing on prognosis of patients with acute subdural hematoma. Chin J Traumatol. Oct/2009. 12:296-8.
  162. Croce MA, Dent DL, Menke PG, et al. Acute subdural hematoma: nonsurgical management of selected patients. J Trauma. 1994 Jun. 36(6):820-6; discussion 826-7. [View Abstract]
  163. Orlando A, Levy AS, Rubin BA, Tanner A, Carrick MM, Lieser M, et al. Isolated subdural hematomas in mild traumatic brain injury. Part 1: the association between radiographic characteristics and neurosurgical intervention. J Neurosurg. 2018 Jun 15. 1-10. [View Abstract]
  164. Patel NY, Hoyt DB, Nakaji P, et al. Traumatic brain injury: patterns of failure of nonoperative management. J Trauma. Mar/2000. 48:367-74.
  165. Nahmias J, Doben A, DeBusk G, Winston S, Alouidor R, Kaye T, et al. Mild Traumatic Brain Injuries Can Be Safely Managed without Neurosurgical Consultation: The End of a Neurosurgical "Nonsult". Am Surg. 2018 May 1. 84 (5):652-657. [View Abstract]
  166. Hutchinson PJ, Kolias AG, Timofeev IS, Corteen EA, Critchley G, Sahuquillo J, et al. Trial of Decompressive Craniectomy for Traumatic Intracranial Hypertension. N Engl J Med. 2016 Sep 7. [View Abstract]
  167. Carney N, Totten AM, O'Reilly C, et al. Guidelines for the Management of Severe Traumatic Brain Injury, Fourth Edition. Brain Trauma Foundation. Available at https://braintrauma.org/coma/guidelines. September 2016; Accessed: September 22, 2016.
  168. Giza CC, Kutcher JS, Ashwal S, Barth J, Getchius TS, Gioia GA, et al. Summary of evidence-based guideline update: evaluation and management of concussion in sports: report of the Guideline Development Subcommittee of the American Academy of Neurology. Neurology. 2013 Jun 11. 80 (24):2250-7. [View Abstract]
  169. Updated Mild Traumatic Brain Injury Guideline for Adults. Centers for Disease Control and Prevention. Available at http://www.cdc.gov/traumaticbraininjury/mtbi_guideline.html. January 22, 2016; Accessed: September 24, 2016.
  170. Holder HD, Gruenewald PJ, Ponicki WR, et al. Effect of community-based interventions on high-risk drinking and alcohol-related injuries. JAMA. 2000 Nov 8. 284(18):2341-7. [View Abstract]
  171. Gentilello LM, Rivara FP, Donovan DM, et al. Alcohol interventions in a trauma center as a means of reducing the risk of injury recurrence. Ann Surg. 1999 Oct. 230(4):473-80; discussion 480-3. [View Abstract]
  172. Thompson DC, Rivara FP, Thompson R. Helmets for preventing head and facial injuries in bicyclists. Cochrane Database Syst Rev. 2000. CD001855. [View Abstract]
  173. Heng KW, Lee AH, Zhu S, et al. Helmet use and bicycle-related trauma in patients presenting to an acute hospital in Singapore. Singapore Med J. 2006 May. 47(5):367-72. [View Abstract]
  174. Lee BH, Schofer JL, Koppelman FS. Bicycle safety helmet legislation and bicycle-related non-fatal injuries in California. Accid Anal Prev. 2005 Jan. 37(1):93-102. [View Abstract]
  175. Siegrist M, Freiberger E, Geilhof B, Salb J, Hentschke C, Landendoerfer P, et al. Fall Prevention in a Primary Care Setting. Dtsch Arztebl Int. 2016 May 27. 113 (21):365-72. [View Abstract]
  176. Donders J, Boonstra T. Correlates of invalid neuropsychological test performance after traumatic brain injury. Brain Inj. 2007 Mar. 21(3):319-26. [View Abstract]
  177. Hutchison M, Comper P, Mainwaring L, Richards D. The influence of musculoskeletal injury on cognition: implications for concussion research. Am J Sports Med. 2011 Nov. 39(11):2331-7. [View Abstract]
  178. Dacey RG Jr, Alves WM, Rimel RW, et al. Neurosurgical complications after apparently minor head injury. Assessment of risk in a series of 610 patients. J Neurosurg. 1986 Aug. 65(2):203-10. [View Abstract]
  179. Deb S, Lyons I, Koutzoukis C. Neuropsychiatric sequelae one year after a minor head injury. J Neurol Neurosurg Psychiatry. 1998 Dec. 65(6):899-902. [View Abstract]
  180. Dikmen SS, Corrigan JD, Levin HS, et al. Cognitive Outcome Following Traumatic Brain Injury. J Head Trauma Rehabil. 2009. 24:430-438.
  181. Cantu RC. Second-impact syndrome. Clin Sports Med. 1998 Jan. 17(1):37-44. [View Abstract]
  182. McCrory P, Meeuwisse W, Johnston K, et al. Consensus statement on Concussion in Sport 3rd International Conference on Concussion in Sport held in Zurich, November 2008. Clin J Sport Med. May/2009. 19:185-200.
  183. McCrory PR, Berkovic SF. Second impact syndrome. Neurology. 1998 Mar. 50(3):677-83. [View Abstract]
  184. Mayers L. Return-to-Play Criteria After Athletic Concussion. Arch Neurol. Sept/2008. 65:1158-1161.
  185. McLendon LA, Kralik SF, Grayson PA, Golomb MR. The Controversial Second Impact Syndrome: A Review of the Literature. Pediatr Neurol. 2016 Sep. 62:9-17. [View Abstract]
  186. Asikainen I, Kaste M, Sarna S. Early and late posttraumatic seizures in traumatic brain injury rehabilitation patients: brain injury factors causing late seizures and influence of seizures on long-term outcome. Epilepsia. 1999 May. 40(5):584-9. [View Abstract]
  187. Angeleri F, Majkowski J, Cacchio G, et al. Posttraumatic epilepsy risk factors: one-year prospective study after head injury. Epilepsia. 1999 Sep. 40(9):1222-30. [View Abstract]
  188. Diaz-Arrastia R, Agostini MA, Frol AB, et al. Neurophysiologic and neuroradiologic features of intractable epilepsy after traumatic brain injury in adults. Arch Neurol. 2000 Nov. 57(11):1611-6. [View Abstract]
  189. Obermann M, Holbe D, Katsarava Z. Post-traumatic headache. Expert Rev Neurother. Sep/2009. 9:1361-1370.
  190. Packard RC, Ham LP. Pathogenesis of posttraumatic headache and migraine: a common headache pathway?. Headache. 1997 Mar. 37(3):142-52. [View Abstract]
  191. Warner JS. Posttraumatic headache--a myth?. Arch Neurol. 2000 Dec. 57(12):1778-80; discussion 1780-1. [View Abstract]
  192. Jorge RE, Robinson RG, Moser D, Tateno A, Crespo-Facorro B, Arndt S. Major depression following traumatic brain injury. Arch Gen Psychiatry. 2004 Jan. 61(1):42-50. [View Abstract]
  193. Singh R, Mason S, Lecky F, Dawson J. Prevalence of depression after TBI in a prospective cohort: The SHEFBIT study. Brain Inj. 2018. 32 (1):84-90. [View Abstract]
  194. Bilgic B, Baral-Kulaksizoglu I, Hanagasi H, et al. Obsessive-compulsive disorder secondary to bilateral frontal damage due to a closed head injury. Cogn Behav Neurol. 2004 Jun. 17(2):118-20. [View Abstract]
  195. Sachdev P, Smith JS, Cathcart S. Schizophrenia-like psychosis following traumatic brain injury: a chart- based descriptive and case-control study. Psychol Med. 2001 Feb. 31(2):231-9. [View Abstract]
  196. McCartan DP, Fleming FJ, Motherway C, Grace PA. Management and outcome in patients following head injury admitted to an Irish Regional Hospital. Brain Inj. 2008 Apr. 22(4):305-12. [View Abstract]
  197. van der Naalt J, van Zomeren AH, Sluiter WJ, et al. One year outcome in mild to moderate head injury: the predictive value of acute injury characteristics related to complaints and return to work. J Neurol Neurosurg Psychiatry. 1999 Feb. 66(2):207-13. [View Abstract]
  198. Chamelian L, Feinstein A. Outcome after mild to moderate traumatic brain injury: the role of dizziness. Arch Phys Med Rehabil. 2004 Oct. 85(10):1662-6. [View Abstract]
  199. DiSanto D, Kumar RG, Juengst SB, Hart T, O'Neil-Pirozzi TM, Zasler ND, et al. Employment Stability in the First 5 Years After Moderate-to-Severe Traumatic Brain Injury. Arch Phys Med Rehabil. 2018 Jul 26. [View Abstract]
  200. Stromberg KA, Agyemang AA, Graham KM, Walker WC, Sima AP, Marwitz JH, et al. Using Decision Tree Methodology to Predict Employment After Moderate to Severe Traumatic Brain Injury. J Head Trauma Rehabil. 2018 Sep 18. [View Abstract]
  201. Harrison-Felix C, Pretz C, Hammond FM, Cuthbert JP, Bell J, Corrigan J, et al. Life Expectancy after Inpatient Rehabilitation for Traumatic Brain Injury in the United States. J Neurotrauma. 2015 Dec 1. 32 (23):1893-901. [View Abstract]
  202. Richmond R, Aldaghlas TA, Burke C, Rizzo AG, Griffen M, Pullarkat R. Age: Is It All in the Head? Factors Influencing Mortality in Elderly Patients With Head Injuries. J Trauma. 2011 Feb 17. [View Abstract]
  203. Ritchie PD, Cameron PA, Ugoni AM, et al. A study of the functional outcome and mortality in elderly patients with head injuries. J Clin Neurosci. 2000 Jul. 7(4):301-4. [View Abstract]
  204. Røe C, Skandsen T, Manskow U, Ader T, Anke A. Mortality and One-Year Functional Outcome in Elderly and Very Old Patients with Severe Traumatic Brain Injuries: Observed and Predicted. Behav Neurol. 2015. 2015:845491. [View Abstract]
  205. Wade DT, King NS, Wenden FJ, et al. Routine follow up after head injury: a second randomised controlled trial. J Neurol Neurosurg Psychiatry. 1998 Aug. 65(2):177-83. [View Abstract]
  206. Cox AL, Coles AJ, Nortje J, et al. An investigation of auto-reactivity after head injury. J Neuroimmunol. 2006 May. 174(1-2):180-6. [View Abstract]
  207. Thomas KE, Stevens JA, Sarmiento K, Wald MM. Fall-related traumatic brain injury deaths and hospitalizations among older adults--United States, 2005. J Safety Res. 2008. 39(3):269-72. [View Abstract]
  208. Jennett B. Epidemiology of head injury. J Neurol Neurosurg Psychiatry. 1996 Apr. 60(4):362-9. [View Abstract]
  209. Wrightson P, Gronwall D. Mild head injury in New Zealand: incidence of injury and persisting symptoms. N Z Med J. 1998 Mar 27. 111(1062):99-101. [View Abstract]
  210. Schanke AK, Sundet K. Comprehensive driving assessment: neuropsychological testing and on- road evaluation of brain injured patients. Scand J Psychol. 2000 Jun. 41(2):113-21. [View Abstract]
  211. Anderson P. Hemodynamic Complications Common in Traumatic Brain Injury. Available at http://www.medscape.com/viewarticle/778999. Accessed: March 25, 2013.
  212. Bazarian JJ, McClung J, Shah MN, et al. Mild traumatic brain injury in the United States, 1998--2000. Brain Inj. 2005 Feb. 19(2):85-91. [View Abstract]
  213. Black K, Zafonte R, Millis S, et al. Sitting balance following brain injury: does it predict outcome?. Brain Inj. 2000 Feb. 14(2):141-52. [View Abstract]
  214. Blumbergs PC, Scott G, Manavis J, et al. Staining of amyloid precursor protein to study axonal damage in mild head injury. Lancet. 1994 Oct 15. 344(8929):1055-6. [View Abstract]
  215. Bruce DA, Alavi A, Bilaniuk L, et al. Diffuse cerebral swelling following head injuries in children: the syndrome of "malignant brain edema". J Neurosurg. 1981 Feb. 54(2):170-8. [View Abstract]
  216. Cassidy JD, Carroll LJ, Cote P, et al. Effect of eliminating compensation for pain and suffering on the outcome of insurance claims for whiplash injury. N Engl J Med. 2000 Apr 20. 342(16):1179-86. [View Abstract]
  217. Centers for Disease Control and Prevention. Sports-related recurrent brain injuries--United States. MMWR Morb Mortal Wkly Rep. 1997 Mar 14. 46(10):224-7. [View Abstract]
  218. Cernak I, Savic VJ, Kotur J, et al. Characterization of plasma magnesium concentration and oxidative stress following graded traumatic brain injury in humans. J Neurotrauma. 2000 Jan. 17(1):53-68. [View Abstract]
  219. Chesnut RM. Intracranial pressure monitoring in brain-injured patients is associated with worsening of survival. J Trauma. Aug/2008. 65:500-1.
  220. Chesnut RM, Marshall LF, Klauber MR, et al. The role of secondary brain injury in determining outcome from severe head injury. J Trauma. 1993 Feb. 34(2):216-22. [View Abstract]
  221. Chiu WT, Kuo CY, Hung CC, et al. The effect of the Taiwan motorcycle helmet use law on head injuries. Am J Public Health. 2000 May. 90(5):793-6. [View Abstract]
  222. Cho YW, Jang SH, Lee ZI, et al. Effect and appropriate restriction period of constraint-induced movement therapy in hemiparetic patients with brain injury: a brief report. NeuroRehabilitation. 2005. 20(2):71-4. [View Abstract]
  223. Collins MW, Grindel SH, Lovell MR, et al. Relationship between concussion and neuropsychological performance in college football players. JAMA. 1999 Sep 8. 282(10):964-70. [View Abstract]
  224. Dharap SB, Khandkar AA, Pandey A, Sharma AK. Repeat CT scan in closed head injury. Injury. 2005 Mar. 36(3):412-6. [View Abstract]
  225. Drake AI, Gray N, Yoder S, et al. Factors predicting return to work following mild traumatic brain injury: a discriminant analysis. J Head Trauma Rehabil. 2000 Oct. 15(5):1103-12. [View Abstract]
  226. Eisenberg HM, Frankowski RF, Contant CF, et al. High-dose barbiturate control of elevated intracranial pressure in patients with severe head injury. J Neurosurg. 1988 Jul. 69(1):15-23. [View Abstract]
  227. Fabbri A, Servadei F, Marchesini G, et al. Early predictors of unfavorable outcome in subjects with moderate head injury in the emergency department. J Neurol Neurosurg Psychiatry. May/2008. 79:567-73.
  228. Feldman Z, Gurevitch B, Artru AA, et al. Effect of magnesium given 1 hour after head trauma on brain edema and neurological outcome. J Neurosurg. 1996 Jul. 85(1):131-7. [View Abstract]
  229. Gordon WA, Brown M, Sliwinski M, et al. The enigma of "hidden" traumatic brain injury. J Head Trauma Rehabil. 1998 Dec. 13(6):39-56. [View Abstract]
  230. Guerra WK, Gaab MR, Dietz H, et al. Surgical decompression for traumatic brain swelling: indications and results. J Neurosurg. 1999 Feb. 90(2):187-96. [View Abstract]
  231. Gusmao SN, Pittella JE. Extradural haematoma and diffuse axonal injury in victims of fatal road traffic accidents. Br J Neurosurg. 1998 Apr. 12(2):123-6. [View Abstract]
  232. Haig AJ, Ruess JM. Recovery from vegetative state of six months'' duration associated with Sinemet (levodopa/carbidopa). Arch Phys Med Rehabil. 1990 Dec. 71(13):1081-3. [View Abstract]
  233. Hall JR, Reyes HM, Horvat M, et al. The mortality of childhood falls. J Trauma. 1989 Sep. 29(9):1273-5. [View Abstract]
  234. Haltiner AM, Newell DW, Temkin NR, et al. Side effects and mortality associated with use of phenytoin for early posttraumatic seizure prophylaxis. J Neurosurg. 1999 Oct. 91(4):588-92. [View Abstract]
  235. Hamill RW, Woolf PD, McDonald JV, et al. Catecholamines predict outcome in traumatic brain injury. Ann Neurol. 1987 May. 21(5):438-43. [View Abstract]
  236. Hanlon RE, Demery JA, Martinovich Z, et al. Effects of acute injury characteristics on neurophysical status and vocational outcome following mild traumatic brain injury. Brain Inj. 1999 Nov. 13(11):873-87. [View Abstract]
  237. Harders A, Kakarieka A, Braakman R. Traumatic subarachnoid hemorrhage and its treatment with nimodipine. German tSAH Study Group. J Neurosurg. 1996 Jul. 85(1):82-9. [View Abstract]
  238. Homayoun P, Parkins NE, Soblosky J, et al. Cortical impact injury in rats promotes a rapid and sustained increase in polyunsaturated free fatty acids and diacylglycerols. Neurochem Res. 2000 Feb. 25(2):269-76. [View Abstract]
  239. Ikonomidou C, Stefovska V, Turski L. Neuronal death enhanced by N-methyl-D-aspartate antagonists. Proc Natl Acad Sci U S A. 2000 Nov 7. 97(23):12885-90. [View Abstract]
  240. Inamasu J, Hori S, Aoki K, et al. CT scans essential after posttraumatic loss of consciousness. Am J Emerg Med. 2000 Nov. 18(7):810-1. [View Abstract]
  241. Jordan BD, Relkin NR, Ravdin LD, et al. Apolipoprotein E epsilon4 associated with chronic traumatic brain injury in boxing. JAMA. 1997 Jul 9. 278(2):136-40. [View Abstract]
  242. Keenan HT, Brundage SI, Thompson DC, et al. Does the face protect the brain? A case-control study of traumatic brain injury and facial fractures. Arch Surg. 1999 Jan. 134(1):14-7. [View Abstract]
  243. Knoller N, Levi L, Shoshan I, et al. Dexanabinol (HU-211) in the treatment of severe closed head injury: a randomized, placebo-controlled, phase II clinical trial. Crit Care Med. 2002 Mar. 30(3):548-54. [View Abstract]
  244. Kobori N, Clifton GL, Dash P, et al. Altered expression of novel genes in the cerebral cortex following experimental brain injury. Brain Res Mol Brain Res. 2002 Aug 15. 104(2):148-58. [View Abstract]
  245. Kobrine AI, Timmins E, Rajjoub RK, et al. Demonstration of massive traumatic brain swelling within 20 minutes after injury. Case report. J Neurosurg. 1977 Feb. 46(2):256-8. [View Abstract]
  246. Lal S, Merbtiz CP, Grip JC. Modification of function in head-injured patients with Sinemet. Brain Inj. 1988 Jul-Sep. 2(3):225-33. [View Abstract]
  247. Landau WM. Tizanidine and spasticity. Neurology. 1995 Dec. 45(12):2295-6. [View Abstract]
  248. Landy PJ. Neurological sequelae of minor head and neck injuries. Injury. 1998 Apr. 29(3):199-206. [View Abstract]
  249. Lane PL, Skoretz TG, Doig G, et al. Intracranial pressure monitoring and outcomes after traumatic brain injury. Can J Surg. 2000 Dec. 43(6):442-8. [View Abstract]
  250. Lang DA, Teasdale GM, Macpherson P, et al. Diffuse brain swelling after head injury: more often malignant in adults than children?. J Neurosurg. 1994 Apr. 80(4):675-80. [View Abstract]
  251. Levin HS, Gary HE Jr, Eisenberg HM, et al. Neurobehavioral outcome 1 year after severe head injury. Experience of the Traumatic Coma Data Bank. J Neurosurg. 1990 Nov. 73(5):699-709. [View Abstract]
  252. Levin HS, Mattis S, Ruff RM, et al. Neurobehavioral outcome following minor head injury: a three-center study. J Neurosurg. 1987 Feb. 66(2):234-43. [View Abstract]
  253. Levin HS, Williams DH, Eisenberg HM, et al. Serial MRI and neurobehavioural findings after mild to moderate closed head injury. J Neurol Neurosurg Psychiatry. 1992 Apr. 55(4):255-62. [View Abstract]
  254. Li J, Brown J, Levine M. Mild head injury, anticoagulants, and risk of intracranial injury. Lancet. 2001 Mar 10. 357(9258):771-2. [View Abstract]
  255. Macfarlane DP, Nicoll JA, Smith C, et al. APOE epsilon4 allele and amyloid beta-protein deposition in long term survivors of head injury. Neuroreport. 1999 Dec 16. 10(18):3945-8. [View Abstract]
  256. Marion DW, Penrod LE, Kelsey SF, et al. Treatment of traumatic brain injury with moderate hypothermia. N Engl J Med. 1997 Feb 20. 336(8):540-6. [View Abstract]
  257. Marmarou A, Anderson RL, Ward JD, et al. Impact of ICP instability and hypotension on outcome in patients with severe head trauma. J Neurosurg. 1991. 75:S59-66.
  258. Marshall LF, Gautille T, Klauber MR, et al. The outcome of severe head injury. J Neurosurg. 1991. 75:S28-36.
  259. Mayers l. Return-to-Play Criteria after Athletic Concussion. Archives of Neurology. Sep/2008. 65:1158-1161.
  260. Miller EC, Derlet RW, Kinser D. Minor head trauma: Is computed tomography always necessary?. Ann Emerg Med. 1996 Mar. 27(3):290-4. [View Abstract]
  261. Mittenberg W, Strauman S. Diagnosis of mild head injury and the postconcussion syndrome. J Head Trauma Rehabil. 2000 Apr. 15(2):783-91. [View Abstract]
  262. Mosimann UP, Muri RM, Felblinger J, et al. Saccadic eye movement disturbances in whiplash patients with persistent complaints. Brain. 2000 Apr. 123 ( Pt 4):828-35. [View Abstract]
  263. Murray JA, Demetriades D, Berne TV, et al. Prehospital intubation in patients with severe head injury. J Trauma. 2000 Dec. 49(6):1065-70. [View Abstract]
  264. Nagy KK, Joseph KT, Krosner SM, et al. The utility of head computed tomography after minimal head injury. J Trauma. 1999 Feb. 46(2):268-70. [View Abstract]
  265. Owings JT, Wisner DH, Battistella FD, et al. Isolated transient loss of consciousness is an indicator of significant injury. Arch Surg. 1998 Sep. 133(9):941-6. [View Abstract]
  266. Packard RC, Ham LP. Posttraumatic headache. J Neuropsychiatry Clin Neurosci. 1994 Summer. 6(3):229-36. [View Abstract]
  267. Palmer AM, Marion DW, Botscheller ML, et al. Traumatic brain injury-induced excitotoxicity assessed in a controlled cortical impact model. J Neurochem. 1993 Dec. 61(6):2015-24. [View Abstract]
  268. Patel NY, Hoyt DB, Nakaji P, et al. Traumatic brain injury: patterns of failure of nonoperative management. J Trauma. 2000 Mar. 48(3):367-74; discussion 374-5. [View Abstract]
  269. Paterakis K, Karantanas AH, Komnos A, et al. Outcome of patients with diffuse axonal injury: the significance and prognostic value of MRI in the acute phase. J Trauma. 2000 Dec. 49(6):1071-5. [View Abstract]
  270. Pop E. Dexanabinol Pharmos. Curr Opin Investig Drugs. 2000 Dec. 1(4):494-503. [View Abstract]
  271. Procaccio F, Stocchetti N, Citerio G, et al. Guidelines for the treatment of adults with severe head trauma (part II). Criteria for medical treatment. J Neurosurg Sci. 2000 Mar. 44(1):11-8. [View Abstract]
  272. Report of the Quality Standards Subcommittee, American Academy of Neurology. Practice parameter: the management of concussion in sports (summary statement). Report of the Quality Standards Subcommittee. Neurology. 1997 Mar. 48(3):581-5. [View Abstract]
  273. Rugg-Gunn FJ, Symms MR, Barker GJ, et al. Diffusion imaging shows abnormalities after blunt head trauma when conventional magnetic resonance imaging is normal. J Neurol Neurosurg Psychiatry. 2001 Apr. 70(4):530-3. [View Abstract]
  274. Salmond CH, Menon DK, Chatfield DA, et al. Diffusion tensor imaging in chronic head injury survivors: correlations with learning and memory indices. Neuroimage. 2006 Jan 1. 29(1):117-24. [View Abstract]
  275. Signoretti S, Marmarou A, Tavazzi B, et al. The protective effect of cyclosporin A upon N-acetylaspartate and mitochondrial dysfunction following experimental diffuse traumatic brain injury. J Neurotrauma. 2004 Sep. 21(9):1154-67. [View Abstract]
  276. Sloan RL, Brown KW, Pentland B. Fluoxetine as a treatment for emotional lability after brain injury. Brain Inj. 1992 Jul-Aug. 6(4):315-9. [View Abstract]
  277. Snoek JW, Minderhoud JM, Wilmink JT. Delayed deterioration following mild head injury in children. Brain. 1984 Mar. 107 ( Pt 1):15-36. [View Abstract]
  278. Speech TJ, Rao SM, Osmon DC, et al. A double-blind controlled study of methylphenidate treatment in closed head injury. Brain Inj. 1993 Jul-Aug. 7(4):333-8. [View Abstract]
  279. Struchen MA, Hannay HJ, Contant CF, et al. The relation between acute physiological variables and outcome on the Glasgow Outcome Scale and Disability Rating Scale following severe traumatic brain injury. J Neurotrauma. 2001 Feb. 18(2):115-25. [View Abstract]
  280. Sturmi JE, Smith C, Lombardo JA. Mild brain trauma in sports. Diagnosis and treatment guidelines. Sports Med. 1998 Jun. 25(6):351-8. [View Abstract]
  281. Sullivan PG, Rabchevsky AG, Waldmeier PC, Springer JE. Mitochondrial permeability transition in CNS trauma: cause or effect of neuronal cell death?. J Neurosci Res. 2005 Jan 1-15. 79(1-2):231-9. [View Abstract]
  282. Teasdale GM. Head injury. J Neurol Neurosurg Psychiatry. 1995 May. 58(5):526-39. [View Abstract]
  283. The Brain Trauma Foundation, The American Association of Neurological Surgeons, The Joint Section on Neurotrauma and Critical Care. Role of steroids. J Neurotrauma. 2000 Jun-Jul. 17(6-7):531-5. [View Abstract]
  284. The Brain Trauma Foundation, The American Association of Neurological Surgeons, The Joint Section on Neurotrauma and Critical Care. Use of barbiturates in the control of intracranial hypertension. J Neurotrauma. 2000 Jun-Jul. 17(6-7):527-30. [View Abstract]
  285. Thurman DJ, Alverson C, Dunn KA, et al. Traumatic brain injury in the United States: A public health perspective. J Head Trauma Rehabil. 1999 Dec. 14(6):602-15. [View Abstract]
  286. Towne AR, Waterhouse EJ, Boggs JG, et al. Prevalence of nonconvulsive status epilepticus in comatose patients. Neurology. 2000 Jan 25. 54(2):340-5. [View Abstract]
  287. van Reekum R, Cohen T, Wong J. Can traumatic brain injury cause psychiatric disorders?. J Neuropsychiatry Clin Neurosci. 2000 Summer. 12(3):316-27. [View Abstract]
  288. Velmahos GC, Jindal A, Chan LS, et al. "Insignificant" mechanism of injury: not to be taken lightly. J Am Coll Surg. 2001 Feb. 192(2):147-52. [View Abstract]
  289. Wesson D, Spence L, Hu X, et al. Trends in bicycling-related head injuries in children after implementation of a community-based bike helmet campaign. J Pediatr Surg. 2000 May. 35(5):688-9. [View Abstract]
  290. Whelan FJ, Walker MS, Schultz SK. Donepezil in the treatment of cognitive dysfunction associated with traumatic brain injury. Ann Clin Psychiatry. 2000 Sep. 12(3):131-5. [View Abstract]
  291. Whyte J, Hart T, Schuster K, et al. Effects of methylphenidate on attentional function after traumatic brain injury. A randomized, placebo-controlled trial. Am J Phys Med Rehabil. 1997 Nov-Dec. 76(6):440-50. [View Abstract]
  292. Wilberger JE Jr, Harris M, Diamond DL. Acute subdural hematoma: morbidity, mortality, and operative timing. J Neurosurg. 1991 Feb. 74(2):212-8. [View Abstract]
  293. Withaar FK, Brouwer WH, van Zomeren AH. Fitness to drive in older drivers with cognitive impairment. J Int Neuropsychol Soc. 2000 May. 6(4):480-90. [View Abstract]
  294. Young B, Runge JW, Waxman KS, et al. Effects of pegorgotein on neurologic outcome of patients with severe head injury. A multicenter, randomized controlled trial. JAMA. 1996 Aug 21. 276(7):538-43. [View Abstract]
  295. Zafonte RD, Mann NR. Cerebral salt wasting syndrome in brain injury patients: a potential cause of hyponatremia. Arch Phys Med Rehabil. 1997 May. 78(5):540-2. [View Abstract]
  296. Orlando A, Levy AS, Rubin BA, Tanner A, Carrick MM, Lieser M, et al. Isolated subdural hematomas in mild traumatic brain injury. Part 1: the association between radiographic characteristics and neurosurgical intervention. J Neurosurg. 2018 Jun 15. 1-10. [View Abstract]
  297. Leland A, Tavakol K, Scholten J, Libin AV, Mathis D, Maron D, et al. Affective and Cognitive Conditions are Stronger Predictors of Success with Community Reintegration than Gait and Balance Performance in Veterans with Mild Traumatic Brain Injury. Med Arch. 2017 Dec. 71 (6):417-423. [View Abstract]

This 50-year-old woman with epilepsy seized and struck her head. Her initial Glasgow Coma Scale score was 12. Her scan shows prominent right temporal bleeding. She recovered to baseline without surgery.

This 50-year-old woman with epilepsy seized and struck her head. Her initial Glasgow Coma Scale score was 12. Her scan shows prominent right temporal bleeding. She recovered to baseline without surgery.

This 40-year-old woman was anticoagulated with warfarin (Coumadin) and fell out of her hospital bed. She subsequently died. Her CT scan shows an obvious right subdural hematoma with mass effect.

This 35-year-old man was in a motor vehicle accident. His initial Glasgow Coma Scale score was 7. He had left hemiparesis. He recovered orientation to temporal parameters after 1 week, but he remained disinhibited and hemiparetic (although able to ambulate). His MRI shows a diffusion-weighted hyperintensity in the right posterior internal capsular limb. This was attributed to an axonal injury. (An embolic workup for stroke was unremarkable, and no dissection was discerned on a carotid Doppler study.)

This 50-year-old woman with epilepsy seized and struck her head. Her initial Glasgow Coma Scale score was 12. Her scan shows prominent right temporal bleeding. She recovered to baseline without surgery.

This 50-year-old man was struck in the head in an assault. His scan shows a right acute subdural hematoma with no mass effect. His initial Glasgow Coma Scale score was 15. He returned home without major sequelae after 5 days of hospitalization.

This is a superior view of the CT scan shown in the previous image. This demonstrates a small left frontal intracranial contusion with some surrounding edema. This could be a marker of axonal injury.

This 23-year-old woman was in a motor vehicle accident with impact on the left. Her initial Glasgow Coma Scale score was 6 and she required intubation. Her scan shows a subtle right posterior frontal linear hyperdensity, most likely a small petechial bleed (contrecoup). This could also be a marker of axonal injury.

This 35-year-old man was in a motor vehicle accident. His initial Glasgow Coma Scale score was 7. He had left hemiparesis. He recovered orientation to temporal parameters after 1 week, but he remained disinhibited and hemiparetic (although able to ambulate). His MRI shows a diffusion-weighted hyperintensity in the right posterior internal capsular limb. This was attributed to an axonal injury. (An embolic workup for stroke was unremarkable, and no dissection was discerned on a carotid Doppler study.)

This 40-year-old woman was anticoagulated with warfarin (Coumadin) and fell out of her hospital bed. She subsequently died. Her CT scan shows an obvious right subdural hematoma with mass effect.

This elderly woman had a history of frequent falls and presented with seizures, possibly from her right hypodense subdural hematoma shown here. The subdural hematoma appears chronic and exhibits no mass effect.

This 23-year-old freelance graphic artist has drifted from job to job following his head injury 2 years prior to this scan. He was hospitalized initially for about 1 week for intracranial bleeding. This CT scan shows obvious medial bifrontal atrophy.