You are on page 1of 13

Stocchetti et al.

Critical Care 2013, 17:201


http://ccforum.com/content/17/1/201

REVIEW

Clinical review: Neuromonitoring - an update


Nino Stocchetti*1, Peter Le Roux2, Paul Vespa3, Mauro Oddo4, Giuseppe Citerio5, Peter J Andrews6, Robert D Stevens7,
Tarek Sharshar8, Fabio S Taccone9 and Jean-Louis Vincent9

Pathophysiology of acute brain injury


Abstract The pathophysiology of acute brain injury is complex and
Critically ill patients are frequently at risk of neurological can involve several secondary pathological cascades that
dysfunction as a result of primary neurological contribute to aggravate neuronal injury (Figure  1). The
conditions or secondary insults. Determining which clinical rationale for neuromonitoring is to tailor therapy
aspects of brain function are affected and how best to patient-specific pathophysiology rather than to pre-
to manage the neurological dysfunction can often be defined thresholds or targets. It is thus important to
difficult and is complicated by the limited information briefly review some basic aspects of brain physiology to
that can be gained from clinical examination in such help understand the techniques and applications of
patients and the effects of therapies, notably sedation, neuromonitoring.
on neurological function. Methods to measure and
monitor brain function have evolved considerably in Cerebral metabolism
recent years and now play an important role in the The human brain constitutes 2% of body weight, yet the
evaluation and management of patients with brain energy-consuming processes that enable adequate brain
injury. Importantly, no single technique is ideal for function account for about 25% of total body energy
all patients and different variables will need to be expenditure and 20% of the oxygen consumption of the
monitored in different patients; in many patients, a whole organism. Glucose is the main energy substrate of
combination of monitoring techniques will be needed. the brain and, given the low glycogen stores in the brain,
Although clinical studies support the physiologic brain glucose levels are highly dependent on blood
feasibility and biologic plausibility of management glucose. Transport of glucose from the systemic circu-
based on information from various monitors, data lation to the brain is a tightly regulated process mediated
supporting this concept from randomized trials are still by specialized cell membrane glucose transporters
required. (GLUT). Experimental and human studies show evidence
of flow-metabolism uncoupling and increased glucose
utilization after acute brain injury (‘cerebral hyper-
Introduction glycolysis’). Importantly, this can occur in the absence of
The overall aims of neuromonitoring are to: 1)  identify low cerebral blood flow (CBF) and cerebral ischemia and
worsening neurological function and secondary cerebral may lead to a state of reduced availability of the main
insults that may benefit from specific treatment(s); energy substrate (glucose) with a subsequent risk of
2) improve pathophysiological understanding of cerebral cerebral energy dysfunction [1,2]. However, hyperglyco-
disease in critical illness; 3)  provide clear physiological lysis also results in increased processing of glucose to
data to guide and individualize therapy; 4)  assist with pyruvate by astrocytes and conversion to lactate.
prognostication. In this article, we will outline the neuro- Endogenous lactate, released in the extracellular space,
monitoring techniques currently in use in critically ill can in turn be transferred, via specific monocarboxylate
patients and suggest how they should be best applied to transporters, to neurons (a process known as the ‘astro-
help us care for such patients. We will focus on clinically cyte-neuron lactate shuttle’) [3]. Brain lactate may thus
available techniques and not discuss new approaches that be used as an alternative aerobic energy substrate to glucose
are still largely in the research stage of development. [4]. Indeed, studies in subarachnoid hemorrhage (SAH)
patients suggest that a pattern of increased cerebral hyper-
*Correspondence: stocchet@policlinico.mi.it
1
Milan University, Terapia Intensiva Neuroscienze, Fondazione IRCCS Ca’ Granda,
glycolytic lactate is associated with better outcome [5].
Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122 Milano, Italy
Full list of author information is available at the end of the article Cerebral blood flow, oxygen delivery and ischemia
Cerebral ischemia is a frequent cause of secondary brain
© 2010 BioMed Central Ltd © 2013 BioMed Central Ltd injury [6]. In normal conditions, reduced systemic
Stocchetti et al. Critical Care 2013, 17:201 Page 2 of 13
http://ccforum.com/content/17/1/201

Assessment Method for the ICU (CAM-ICU) [13] or the


ICU Delirium Screening Checklist (ICU DSC) [14] can
be used to assess the presence and degree of delirium.
Neurological examination should then assess for neck
stiffness, motor responses, plantar and deep tendon
reflexes and cranial nerve functions. The presence of
focal neurological signs should prompt brain imaging,
which should also be considered, along with an electro-
encephalogram (EEG), if there is no obvious cause for
delirium or coma.

Monitoring techniques
Figure 1. Some of the pathological cascades contributing
Intracranial pressure
to aggravate neuronal injury after acute brain injury. Initial Elevated intracranial pressure (ICP; >20 mmHg) is asso-
mechanisms, such as excitotoxicity and mitochondrial dysfunction, ciated with increased mortality after acute traumatic
initiate damage in the very early minutes/hours; other factors, such as brain injury (TBI) [15]. The most recent Brain Trauma
energy dysfunction, edema, and inflammation come into play later in Foundation guidelines [16] recommend (level II
the process. CBF, cerebral blood flow.
evidence) that ICP should be monitored in all salvageable
patients with severe TBI (GCS score of 3 to 8 after
resuscitation) and an abnormal head CT scan.
pressure triggers an active vasodilatory response that ICP monitoring is also used in non-traumatic neuro-
keeps cerebral blood flow (CBF) constant over a wide logical disorders, such as SAH, and to a lesser extent in
range of mean arterial pressures (MAPs), thereby pre- brain tumors, infarctions, intracerebral hemorrhage, and
venting brain hypoperfusion (cerebral pressure autoregu- infections. After SAH, external ventricular drainage is
lation) [7]. In brain-injured patients, cerebrovascular recommended in patients with higher World Federation
reactivity may be impaired, and a decrease in MAP or of Neurological Societies (WFNS) scale scores or acute
cerebral perfusion pressure (CPP) may thus translate into hydrocephalus [17], to monitor ICP and to drain cerebro-
reduced CBF and secondary ischemia [8]. spinal fluid (CSF) for ICP control. In spontaneous intra-
Cerebral oxygen utilization is proportional to the cerebral hemorrhage, ICP monitoring is seldom used, but
product of CBF and the arterio-venous difference in recent guidelines [18] suggest that in patients with a GCS
oxygen content [9]. Mechanisms other than perfusion- score of <8, those with clinical evidence of transtentorial
limited ischemia (microvascular collapse, endothelial herniation, or those with intraventricular hemorrhage or
swelling, perivascular edema) may increase the diffusion hydrocephalus, ICP monitoring and treatment may be
gradient for oxygen between venous blood and brain considered (class IIb; level of evidence: C).
tissue (PvO2-PbtO2), reduce cellular oxygen delivery and ICP is a function of intracranial content: some content
attenuate the ability of the brain to increase oxygen is easy to detect and correct, such as an expanding
extraction fraction in response to reduced CBF [10]. contusion, while others are more difficult - for example,
in cases of diffuse cerebral swelling, which may indicate
Neurological examination of the ICU patient vasodilation or edema. The volume-pressure curve,
Clinical neurological examination is a fundamental com- which describes ICP changes to corresponding increases
ponent of neuromonitoring and should take into account in volume, has an exponential shape: as long as com-
the effects of sedative drugs, which may markedly pensatory mechanisms can be used (that is, displacement
influence neurological responses. The degree of aware- of CSF from the supratentorial space into the spinal sac),
ness should first be assessed. Clinical assessment is based ICP will remain constant despite the volume increase.
mainly on evaluating eye and motor responses to verbal When compensatory capabilities are exceeded, ICP rises
orders and noxious stimuli. The depth of coma can be sharply. Attempts have been made to identify loss of
evaluated by the Glasgow Coma Scale (GCS) [11] or the compensation using ICP waveform analysis: the normal
Full Outline of UnResponsiveness (FOUR) score [12], ICP waveform has three components (P1 > P2 > P3), but
which includes assessment of the pupillary light and is altered when pressure increases, with an increase in
corneal reflexes and of the breathing pattern. Delirium is the P2 component compared to P1 (Figure  2); the ICP
a fluctuating state characterized by altered attention, pulse amplitude (difference between systolic and diastolic
spatio-temporal disorientation, disorganized thinking values) also tends to increase [19]. However, this feature
and alteration of awareness. Delirious patients can be depends on the CPP and on the physical properties of the
hypoactive, hyperactive or both alternately. The Confusion measuring system, and so should be used with caution.
Stocchetti et al. Critical Care 2013, 17:201 Page 3 of 13
http://ccforum.com/content/17/1/201

Figure 2. Intracranial pressure curve in conditions of normal and reduced compliance. Valuable information on intracranial buffering
mechanisms can be extracted from the shape of the curves: when all normal components (peaks) of the intracranial pressure (ICP) curve are clearly
evident (left panel), the intracranial system is behaving normally and small increases in volume can still be compensated for. However, when a P2-
predominant pattern is displayed (right panel), the intracranial system is less compliant and compensatory abilities are reduced.

Although non-invasive evaluation of ICP is possible TDP monitoring may also guide CPP management after
using the transcranial Doppler (TCD)-derived pulsatility TBI [25]. TDP data are currently limited to small single-
index [20] or optic nerve sonography [21], the only center studies and larger trials are needed to confirm the
methods for continuous on-line monitoring of ICP validity and use of this technique.
remain invasive. Intra-ventricular devices have long been
considered the ‘gold standard’; however, intra-paren- Transcranial Doppler
chymal pressure monitoring provides equivalent pressure Direct measurement of blood flow in the brain arteries
measurements [22]. Intraventricular probes are particu- would be of value in all cases of brain damage. Using an
larly useful when CSF drainage is desirable. ultrasound probe, flow velocity (rather than flow itself )
ICP management should also take CPP into account can be measured in the main arteries of the base of the
(Figure 3). Therapies to reduce intracranial hypertension brain. TCD combines ultrasound and the Doppler
that also may reduce arterial pressure and CPP (for principle to represent erythrocyte flow in the basal
example, barbiturates) require careful titration to preserve cerebral arteries. In some 10% of patients, transtemporal
adequate cerebral perfusion. insonation is not feasible because of anatomical barriers.
When cerebrovascular resistance increases - for example,
Cerebral blood flow in vasospasm of the middle cerebral artery - systolic TCD
Direct, continuous measurement of regional CBF is now velocity increases, whereas diastolic velocity decreases,
feasible using a thermal diffusion probe (TDP; Hemedex® leading to a clear increase in the pulsatility index (the
Cambridge, Massachusetts, USA) inserted into the brain ratio of the difference between systolic and diastolic flow
parenchyma. The sensor at the tip provides a quantitative to diastolic flow). For this reason, TCD is often used to
measure in the volume of tissue surrounding the sensor. monitor the time course of vasospasm after SAH [26].
The TDP technique showed good agreement with CBF However, correct interpretation of the pulsatility index is
measured by xenon-CT [23]. complex, because it depends not only on cerebrovascular
Correct and stable positioning of regional CBF probes resistance, but also on several systemic and cerebral
with a skull bolt is crucial and a CT-scan should be variables [27].
performed to verify correct placement. Precise quanti- TCD has also been used to assess cerebrovascular
fication of regional CBF using a TDP is dependent on the autoregulation in TBI and SAH patients [7]. Flow velocity
presence of a stable temperature and may be altered in responses to spontaneous or induced (with vasopressors)
conditions of hyperthermia or rapid fluctuation in a changes in CPP may quantify the status of static pressure
patient’s temperature. autoregulation; autoregulation estimated by TCD in the
In patients with SAH, Muench and colleagues [24] used middle cerebral artery correlated with autoregulation
TDP monitoring to guide medical therapy for delayed studied by positron emission tomography in TBI patients
cerebral ischemia, and demonstrated that increasing [28].
blood pressure with vasopressors was the only inter- The main advantage of TCD is that it is non-invasive
vention that improved CBF and PbtO2; hypervolemia and and can be carried out at the bedside. However, the
hemodilution had only marginal effects at best. Hence, quality of the TCD signal is operator-dependent and
normovolemia and MAP/CPP augmentation are increas- correct interpretation requires training. TCD signals may
ingly used instead of ‘triple-H’ therapy in these patients. also vary over time with temperature, arterial carbon
Stocchetti et al. Critical Care 2013, 17:201 Page 4 of 13
http://ccforum.com/content/17/1/201

Figure 3. Intracranial pressure (ICP), mean arterial pressure (MAP), and cerebral perfusion pressure (CPP) in a patient with traumatic brain
injury undergoing bronchoscopy under general anesthesia with myorelaxants. ICP increased to 45 mmHg and CPP decreased concomitantly
to 40 mmHg. Broncoscopy was stopped, further sedation given and manual hyperventilation administered briefly. ICP and CPP returned to normal
levels.

dioxide tension (PaCO2 and, in some series, correlation to coupling of flow to metabolism [31]. Unfortunately, it
with actual CBF measurements was disappointing [29]. is still not possible to visualize the brain microcirculation
in clinical practice without direct exposure of the cerebral
Microcirculation cortex after craniectomy and data on microvascular
Microcirculatory alterations play a key role in the patho- abnormalities in humans are limited.
genesis of organ dysfunction. Brain endothelial cells help Direct evaluation of the brain microcirculation can be
regulate vessel diameter and permeability [30], contributing performed using sidestream dark field imaging and
Stocchetti et al. Critical Care 2013, 17:201 Page 5 of 13
http://ccforum.com/content/17/1/201

intravital microscopy [32]. These methods allow con- flow and metabolism, particularly when CBF is manipu-
tinuous and in vivo observation of the microcirculation at lated - for example, when using hyperventilation.
high resolution as well as evaluation of vessel size,
number, density and flow. Although intravital microscopy Direct PbtO2 measurement
is still considered the gold standard, it is not suitable for Direct PbtO2 monitors are the most common technique
human use. The sidestream dark field technique can be used in the ICU to assess cerebral oxygenation. Probe
applied on exposed cortical areas during craniectomy but positioning is crucial: usually inserted in the white
care must be taken to limit pressure artifacts and matter, readings are dependent, in part, on proximity to
minimize the risk of infection [33]. In patients with intracranial pathology [42]. For example, if close to a
stroke from middle cerebral artery occlusion who under- contusion, values may be reduced and this should be
went decompressive craniectomy, brain microcirculatory considered when making management decisions. When
density and flow were significantly altered when com- the PbtO2 probe is in what appears to be normal white
pared to control patients [34]. Similarly, altered micro- matter on CT, then the reading, although local in nature,
vascular reactivity has been demonstrated in patients provides a reasonable estimate of global brain oxygenation.
with SAH [35]. PbtO2 is not a ‘surrogate’ for ischemia or CBF, because
it varies not only with CBF (and factors that regulate it -
Brain tissue oxygen monitoring for example, CO2 and MAP) but also with changes in
Several techniques can be used to measure brain arterial oxygen tension (PaO2) among many other factors.
oxygenation, the most common in the ICU being jugular PbtO2 is, therefore, more a marker of the balance between
venous bulb oximetry and direct PbtO2 measurement. regional oxygen supply and cellular oxygen consumption
Near infra-red spectroscopy has also been used for this [9].
purpose. Monitoring PbtO2 has been validated against fiberoptic
SjO2 monitoring, xenon-enhanced CT scanning, and
Jugular venous bulb oximetry SPECT. Threshold values vary slightly depending on what
This technique requires placement of a catheter into the type of PbtO2 monitor is used but values <20 mmHg are
jugular bulb. The arterio-jugular difference in oxygen considered worth treating and values <15 mmHg indicate
content (AJDO2) is proportional to CBF and inversely brain hypoxia or ischemia [43,44]. Decreases in PbtO2
proportional to oxygen consumption (cerebral metabolic have been associated with independent chemical markers
rate for oxygen, CMRO2) and is often used as a global of brain ischemia in microdialysis studies [45]. The
measure of adequate perfusion. Proper positioning of the number, duration, and intensity of brain hypoxic episodes
probe in the jugular bulb is crucial, since blood draining (PbtO2 <15 mmHg) and any PbtO2 values ≤5 mmHg are
from extra-cerebral structures, such as the neck and face, associated with poor outcome after TBI [46,47]. Episodes
can contaminate the lower portions of the jugular vein. of brain hypoxia are common and may occur even when
Under most conditions in which arterial hemoglobin ICP and CPP are normal [48]. The exact relationship with
saturation is constant, just the jugular oxygen saturation outcome may, however, vary according to whether the
(SjO2) is measured. This measurement can be performed probe is positioned in normal white matter, the
by intermittent sampling, or continuously using fiber- penumbra or in a contusion [42,49]. Monitoring of PbtO2
optic probes. Normal values for SjO2 in patients without can be used in combination with other intra-parenchymal
brain damage are about 57% (95% confidence interval 52 monitors, mainly ICP and cerebral microdialysis. Clinical
to 62%) [36]. In the early hours after trauma, low SjO2 studies suggest that therapy based on information from
values have been detected, especially in the most severe both an ICP and a PbtO2 monitor may be associated with
cases [37]. Severe and frequent SjO2 desaturations are better outcomes than that based on ICP monitoring
associated with worse outcome in TBI patients [38]. alone [50].
Because of the anatomy of the cerebral circulation, SjO2
is an average indicator and a large volume of brain must Near infra-red spectroscopy
be underperfused for an abnormality to be detected. Oxyhemoglobin, deoxyhemoglobin, and oxidized cyto-
Two-thirds of the blood flowing in each jugular vein chrome oxidase absorb specific portions of the light
comes from the ipsilateral and one-third from the spectra. When a tissue layer is illuminated with a source
contralateral hemisphere [39]; focal areas of mismatch of light in the near infra-red wavelength, the attenuation
between flow/metabolism can, therefore, be missed by of the light signal is correlated to the relative proportions
this global measurement [40]. Additionally, SjO2 values in of oxyhemoglobin and deoxyhemoglobin (HbO2/Hb) and
one jugular vein may differ from values in the other [41]. oxidized cytochrome oxidase in the tissue. Using appro-
Despite these limitations, intermittent SjO2 sampling is a priate calculations, an estimate of tissue oxygenation can,
cheap and relatively easy tool for estimating adequacy of therefore, be made non-invasively and continuously.
Stocchetti et al. Critical Care 2013, 17:201 Page 6 of 13
http://ccforum.com/content/17/1/201

Near infra-red spectroscopy has been used in TBI [51] or severe disability in severe TBI [63,64], and following
and SAH [52], with ambiguous results. The technique has cardiac arrest [65] or aneurysmal SAH [66]. Although
major limitations in adults: baseline normal values vary highly sensitive, the specificity of this marker for acute
widely, extracranial contamination is a problem, and brain injury is generally low [67].
poor correlation with independent measurements of
cerebral oxygenation has been reported frequently. Glial fibrillary acidic protein
Glial fibrillary acidic protein is associated with inter-
Microdialysis mediate filaments in astrocytes. Increased serum levels
Cerebral microdialysis (CMD) involves the insertion of a are observed following moderate and severe TBI [68],
catheter tipped with a semi-permeable membrane ischemic stroke [69], SAH [70], and cardiac arrest [71].
(usually with a 20 kDa cutoff ) in the brain parenchyma. Serum glial fibrillary acidic protein levels more accurately
The CMD catheter is constantly perfused, thereby allow- discriminate TBI outcomes than do levels of s-100β [72].
ing regular sampling of the patient’s brain extracellular
fluid [53]. CMD sampling is limited to the interstitial Neuron-specific enolase
tissue area around the catheter, thus measuring regional Neuron-specific enolase is an enzyme that is found in
brain metabolism. In clinical practice, a pattern of neurons, cells of neuroendocrine origin, erythrocytes and
elevated lactate/pyruvate ratio and low glucose is con- platelets. Increased serum neuron-specific enolase has
sidered as a warning sign for cerebral ischemia/hypoxia been observed in patients with ischemic stroke, TBI,
[54,55]. High lactate/pyruvate ratios in normoxic condi- SAH and after cardiac arrest [63,65,73]. Neuron-specific
tions have been interpreted as markers of hyperglycolysis. enolase can help discriminate between favorable and
Elevated glutamate is a marker of cellular dysfunction, unfavorable outcomes following severe TBI and cardiac
for example, delayed cerebral ischemia in high-risk SAH arrest [63,65].
patients [56]. Absolute CMD values are important, but
trends over time may provide more useful information. Tau
Recently, CMD has been used to identify the ‘optimal’ Tau is a neuronal protein that stabilizes microtubules,
threshold for blood glucose during insulin therapy [57], thereby promoting axonal transport. Significant eleva-
and when used in combination with PbtO2, CMD moni- tions in tau have been observed in the CSF [74] and brain
toring has potential clinical utility to target adequate interstitial fluid [75] of patients with severe TBI. CSF tau
values of MAP/CPP [58,59] and blood hemoglobin protein is also elevated in patients with SAH, and is
concentration [60]. associated with neurological severity and outcome [76].

Biomarkers Amyloid β
Neurological biomarkers, quantitative indicators of brain Amyloid β is a peptide cleaved from neuronal and glial
dysfunction or damage, may be obtained via sampling of amyloid precursor protein. CSF amyloid β fragment
biological tissues (blood, CSF, brain interstitial fluid), levels are lower in TBI and in SAH patients than in
electrophysiological recordings, or neuroimaging. For control subjects, and lower levels are predictive of short-
soluble biomarkers, modeling of extracerebral concentra- term functional outcome [77]. Recently, amyloid β has
tion kinetics is complex and must account for passage been measured in brain interstitium [75,78].
across anatomical barriers into the bloodstream or CSF;
serum levels of a marker may therefore correlate less Neurofilament heavy chain
closely with brain levels than with alterations in blood- Elevated levels of NfH (neurofilament heavy chain), a
brain barrier integrity. An emerging body of work neuronal protein that is integral to the axonal
suggests that multi-marker panels may enhance sensi- cytoskeleton, have been reported in the brain interstitial
tivity and specificity for acute neurological injury [61,62]. fluid of patients with severe TBI and are associated with
There is also considerable interest in genomic, proteomic physiological abnormalities and mortality [79]. CSF NfH
and lipidomic profiles. is increased following SAH and hypertensive intra-
Brain biomarkers are categorized according to their cerebral hemorrhage [80,81]. Serum concentrations of
source as primarily neuronal, astroglial, or microglial. NfH do not reliably identify brain injury, perhaps because
Here we briefly review the key biomarkers that have been the large size of this molecule impedes passage through
tested clinically in acute neurological injury. the blood-brain barrier.

s-100β Ubiquitin carboxy-terminal hydrolase L1


Increased levels of this astroglial protein are associated CSF levels of UCH-L1 (Ubiquitin carboxy-terminal
with injury severity and are predictive of long-term death hydrolase L1) are significantly elevated in severe TBI
Stocchetti et al. Critical Care 2013, 17:201 Page 7 of 13
http://ccforum.com/content/17/1/201

patients and are associated with lower levels of data that have been trended over time. The lack of PAV
consciousness and worse six-month functional outcomes over time during the initial three days after severe TBI
[82]. Serum levels of UCH-L1 are associated with TBI was associated with clinical outcome at 1 month [93] and
severity and survival [83]. 6 months [94]. PAV trends are automatically created by
modern EEG software, and can be useful in tracking
Alpha II-spectrin improvement or lack of improvement in patients in real
Alpha II-spectrin is a fundamental component of the time (Figure 5). Caution is required when using PAV and
neuronal cytoskeleton, which in the setting of injury is other qEEG parameters, however, because they may be
degraded into fragments referred to as spectrin break- affected by deep sedation.
down products. CSF concentrations of these are elevated
after TBI and correlate with the severity of injury and Electrocorticography and spreading depression
with six-month outcome [84]. Spectrin breakdown Electrodes placed on the surface of the human cortex
products are also detectable in serum and have diagnostic (electrocorticography), usually in proximity to damaged
and prognostic value in children with TBI [85]. areas, can detect spontaneous waves of depolarization,
which appear as spreading depression. Evidence is
Electro-physiological measurements accumulating that these spreading depolarization waves
Electroencephalogram cause an imbalance in energy consumption and delivery
The classical indication for EEG is to detect and manage and may lead to secondary brain injury [95,96]. Spreading
seizures, including status epilepticus. In critically ill depression has been identified in TBI [97] and in patients
patients, seizures are often non-convulsive and may with SAH [98] in whom the number of spreading
aggravate brain injury [86]. Intermittent EEG is less depressions correlated significantly with the development
sensitive than continuous EEG for detecting non- of delayed cerebral ischemia even in the absence of
convulsive status epilepticus [87]. Approximately 50% of vasospasm. Electrocorticography requires a craniotomy,
non-convulsive seizures are recorded within the first but recent studies have suggested there may be correlates
60 minutes of EEG recording, but in some patients up to with human scalp direct and alternating current
48 hours of monitoring may be required [88]. Over the electroencephalography [99].
past decade, a more advanced form of EEG, quantitative
EEG (qEEG), has been developed, in which the raw EEG Evoked potentials
signal is converted into a digital form using fast Fourier Somatosensory evoked potentials (SSEPs) are measured
transformation (compressed spectral array) (Figure 4). on the scalp as evoked EEG responses to an electrical
Several groups have used qEEG to detect seizures and stimulus applied typically to the median or tibial nerves.
other causes of brain dysfunction, such as ischemia. In SSEPs are less affected by pharmacological agents or
SAH patients, the percent alpha trend variability (PAV) hypothermia than is the EEG. The main variable used for
decreased acutely with the onset of vasospasm-induced prognosis is the cortical response, which usually occurs
decreases in CBF and returned to normal with treatment at 20 msec after the stimulus, and hence is called the N20
and resolution of vasospasm [89]. More recently, Ratha- peak. After cardiac arrest, bilateral absence of the N20
krishnan and colleagues [90] reported that changes in α SSEP is associated with persistent vegetative state or
power could help to distinguish delayed cerebral ischemia death in all patients [100,101]. SSEPs can be measured
from vasospasm after SAH. Thus, there appears to be during periods of sedation and even hypothermia, but
good evidence for the use of qEEG to detect cerebral most clinicians wait for return of normothermia. There is
ischemia. some concern about inter- and intra-observer variability
qEEG may also be used to monitor the depth of in interpretation of SSEPs [102]. The use of so-called
sedation in the ICU. The most commonly used modality ‘cognitive’ potentials, such as N70, has been suggested to
is the ‘bispectral index’ (BIS) [91], an algorithm-derived improve the predictive value of poor prognosis of this
number that approximates the degree of sedation- test but this approach is not widely used [103].
induced EEG activity when patients are under general
anesthesia. It ranges from 0 (brain dead) to 100 (normal Imaging
brain function); a value of <60 indicates general anes- Computed tomography
thesia, and <30 indicates burst suppression. CT is the imaging modality of choice in the initial
Selected characteristics of raw EEG, such as the back- evaluation of patients with TBI or when acute hemor-
ground rhythm and Synek scale, spontaneous variability rhage is suspected. It helps to rule out surgical masses,
and responsiveness stimulation, have been used to assist and to identify early signs of intracranial hypertension.
in estimating prognosis [92]. qEEG is more objective Since hemorrhagic lesions or edema may evolve over the
because it uses derived measures, and a larger amount of first hours after injury, a CT scan must be repeated
Stocchetti et al. Critical Care 2013, 17:201 Page 8 of 13
http://ccforum.com/content/17/1/201

Figure 4. Transformation of raw electroencephalogram (EEG) into quantitative EEG (qEEG). The center top panel shows a 10 second segment
of raw EEG. This EEG segment can be transformed using mathematical processing to separate the waves into major frequency bins with the height
of each bin indicating the relative percentage (that is, power) of that frequency during the 10 second epoch (lower left panel). Each bandwidth
can be trended on a minute by minute basis to create a variability histogram for that bandwidth for each channel (lower right panel). The alpha
bandwidth variability trend is shown (lower center panel).

whenever there is clinical deterioration even if the initial devices that are incompatible with the magnetic field, the
imaging was apparently normal. The severity of TBI need for sedation or even neuromuscular blockade to
lesions may be classified using the Marshall [104] or prevent movement artifacts, and the risks inherent to
Rotterdam [105] scales. New features, such as angioCT transport outside of the ICU environment.
and CT perfusion, add important information to non- Specific MRI sequences can enhance the diagnostic
contrast CT and are increasingly used in stroke and SAH and prognostic evaluation of patients with acute brain
evaluation. injury.

Magnetic resonance imaging Diffusion weighted imaging


Magnetic resonance imaging (MRI) has great spatial Reductions in the apparent diffusion coefficient (ADC), a
resolution and, in patients with TBI, can identify patho- marker of water diffusion, are associated with acute
logic abnormalities that are undetected or poorly charac- tissue infarction in stroke and in cardiac arrest. Reduced
terized with CT, such as traumatic axonal injury. Acute ADC values have been noted in major gray matter and
ischemic stroke can also be detected earlier using MRI white matter structures after cardiac arrest and are
than CT. MRI is multi-parametric and can provide predictive of six-month outcome [106].
anatomical detail and quantitative information on brain
physiology and metabolism, also allowing neuronal Diffusion tensor imaging
activation to be mapped. MRI has the advantage over Diffusion tensor imaging evaluates the directional
other radiological methods in that it does not require preference (anisotropy) of water diffusion and allows
ionizing radiation. Challenges for MRI scanning of quantitative assessments of white matter damage in
critically ill patients include: monitoring and resuscitation patients with TBI. The magnitude of fractional anisotropy
Stocchetti et al. Critical Care 2013, 17:201 Page 9 of 13
http://ccforum.com/content/17/1/201

Figure 5. Lack of variability over time of the percent alpha trend (PAV) is associated with poor clinical outcomes. The daily PAV scores are
graphed for those brain injured patients who eventually had a good outcome (top panel) and those that had a poor outcome (bottom panel). The
PAV scores tend to remain good or improve day by day in those patients with a good outcome, whereas they remain poor or worsen day by day in
those patients with a bad outcome.

change correlates with clinical TBI severity in the acute susceptibility weighted imaging correlates with neuro-
setting and with long-term outcomes [107,108]. logical severity on initial presentation and with long-term
cognitive impairment and functional disability [109].
Magnetic resonance spectroscopy
Magnetic resonance spectroscopy measures tissue levels Functional MRI
of selected neurometabolites and can provide important Functional MRI demonstrates that cortical-subcortical
insight into TBI pathophysiology and natural history. It networks governing motor activity are significantly
has been demonstrated that N-acetyl aspartate, a marker impaired following TBI [110].
of neuronal integrity, is decreased in TBI, often in brain
tissue that appears normal on cranial CT or on con- Integration of variables
ventional MRI. Brain N-acetyl aspartate levels are A single monitoring technique may not fully describe the
associated with worse clinical condition and are complex pathophysiological changes in the brain and the
predictive of functional outcome following TBI [108]. concept of multimodality monitoring, the simultaneous
digital recording of multiple parameters of brain func-
Susceptibility weighted imaging tion, has been introduced [111]. Multicenter collabora-
Susceptibility weighted imaging is highly sensitive to tions, such as BrainIT (Brain monitoring with Information
microscopic hemorrhagic lesions associated with TBI, Technology collaborative network) [112], have demon-
which are believed to represent a subtype of diffuse strated that recording of many physiological variables
axonal injury. In children with diffuse axonal injury, the across multiple patients is feasible and can provide new
number and volume of hemorrhagic lesions seen with clinical insights [113]. Advanced statistical and
Stocchetti et al. Critical Care 2013, 17:201 Page 10 of 13
http://ccforum.com/content/17/1/201

mathematical tools can be applied to the large volumes of that may benefit from specific treatments; improve
data obtained with the aim of identifying patterns of pathophysiological understanding of cerebral disease
brain injury and providing clinicians with easier in critical illness; provide physiological data to guide
identification of specific targets [114]. Challenges in data and individualize therapy; assist in prognostication.
integration and synchronization remain, but, ultimately, • At present there is no ‘ideal’ single brain monitor; a
this concept should provide real-time, user-friendly combination of monitoring techniques may provide
advanced data analysis that, when applied at the bedside, better insight into brain function than a single monitor
could improve treatment. used alone.
• Trends over time and threshold values are both
Future trials important when assessing brain function.
Neurological monitoring has developed rapidly over the • Clinical studies suggest the physiological feasibility
past 25  years with widespread application of various and biological plausibility of management based on
monitors, but, although clinical studies suggest the information from various monitors but data
physiological feasibility of this concept, there are still no supporting this concept from randomized trials are
published data from randomized trials to support that still required.
targeting any variables improves clinical outcome.
Abbreviations
Neurocritical care research over the past 20  years has CBF, cerebral blood flow; CMD, cerebral microdialysis; CPP, cerebral
focused on the possibility that early pharmacological perfusion pressure; CSF, cerebrospinal fluid; CT, computed tomography;
intervention might improve outcome, which has deflected EEG, electroencephalogram; GCS, Glasgow Coma Scale; ICP, intracranial
pressure; MAP, mean arterial pressure; MRI, magnetic resonance imaging;
attention from trials designed to answer simple questions PAV, percent α trend variability; PbtO2, brain tissue PO2; qEEG, quantitative
about the everyday management of acute brain injury EEG; SAH, subarachnoid hemorrhage; SjO2, jugular oxygen saturation; SSEP,
patients, such as optimum hemoglobin transfusion somatosensory evoked potential; TBI, traumatic brain injury; TCD, transcranial
Doppler; TDP, thermal diffusion probe.
thresholds, hyperventilation, and arterial pressure
management after hemorrhagic stroke, for example. Competing interests
More extensive availability and use of neuromonitoring PLeR has received research funding from Integra and Neurologica and
has received consultancy fees from Codman. PV is an advisor for Edge
technology should facilitate the development of large Pharmaceuticals, Intouch Health, and GE, has received grants from NIH, and
clinical trials networks to provide the infrastructure has equity in Intouch Health. MO is supported by Grants from the Swiss
necessary to design and conduct studies to resolve some National Science Foundation (grant nr. 320030_133103) and the European
Critical Care Research Network (ECCRN). He is a member of the Speakers’
of these issues. bureau of Integra Neuroscience. GC has been involved in educational activities
for Codman Italy and Codman Europe (Speakers’ bureau). PJA has received
Conclusion honoraria from Integra life sciences and BARD/ Medivance. NS, RD, TS, FST and
JLV have no conflicts of interest to declare related to this article.
Monitoring of brain function in critically ill patients must
begin with a careful clinical evaluation. Monitoring Author details
1
systems typically look at single variables and there is a Milan University, Terapia Intensiva Neuroscienze, Fondazione IRCCS Ca’
Granda, Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122 Milano, Italy;
need to integrate/combine systems in order to obtain a 2
Department of Neurosurgery, University of Pennsylvania, 235 S. 8th Street,
full and accurate assessment of the patient’s condition so Philadelphia, PA 19106, USA; 3Department of Neurosurgery, David Geffen
as to be able to select the most appropriate therapy. School of Medicine at UCLA, 757 Westwood Blvd, Suite 6236A, Los Angeles,
CA 90095, USA; 4Department of Critical Care Medicine, CHUV-University
Importantly, neuromonitoring is a dynamic process and Hospital, Faculty of Biology and Medicine, University of Lausanne, Rue du
not a single measurement; the ability to follow changes Bugnon 46, BH 08.623; CH-1011 Lausanne, Switzerland; 5Neuroanestesia
over time is vital to assess response to therapy and e Neurorianimazione, Dipartimento di Medicina Perioperatoria e Terapie
Intensive, H San Gerardo, Via Pergolesi 33, 20052 Monza, Italy; 6Center
predict prognosis. No monitor will, by itself, improve for Clinical Brain Sciences, Critical Care and Pain Medicine, University of
patient outcomes but wise interpretation of the data and Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU,
integration of the information obtained into an UK; 7Division of Neuroscience Critical Care, Johns Hopkins University School
of Medicine, Meyer 8-140, 600 N. Wolfe St, Baltimore, MD 21287, USA;
individualized therapeutic plan should help optimize care 8
Department of Intensive Care Medicine, Raymond Poincaré, Teaching
of critically ill patients with brain injury. Hospital and University of Versailles Saint-Quentin en Yvelines, 104 Boulevard
Raymond Poincaré, 92380 Garches, France; 9Department of Intensive Care,
Erasme Hospital, Université libre de Bruxelles, route de Lennik 808, 1070
Key messages Brussels, Belgium
• Monitoring of brain function should be considered in
all comatose patients in the ICU. Published: 15 January 2013

• Brain monitoring is centered on a careful clinical References


examination, although this can be limited in comatose 1. Glenn TC, Kelly DF, Boscardin WJ, McArthur DL, Vespa P, Oertel M, Hovda DA,
and/or sedated patients. Bergsneider M, Hillered L, Martin NA: Energy dysfunction as a predictor of
outcome after moderate or severe head injury: indices of oxygen, glucose,
• The goals of neuromonitoring are to: identify and lactate metabolism. J Cereb Blood Flow Metab 2003, 23:1239-1250.
worsening neurological function and secondary insults 2. Vespa P, Bergsneider M, Hattori N, Wu HM, Huang SC, Martin NA, Glenn TC,
Stocchetti et al. Critical Care 2013, 17:201 Page 11 of 13
http://ccforum.com/content/17/1/201

McArthur DL, Hovda DA: Metabolic crisis without brain ischemia is R, Manley GT, Nemecek A, Newell DW, Rosenthal G, Schouten J, Shutter L,
common after traumatic brain injury: a combined microdialysis and Timmons SD, Ullman JS, Videtta W, Wilberger JE, Wright DW: Guidelines for
positron emission tomography study. J Cereb Blood Flow Metab 2005, the management of severe traumatic brain injury. VII. Intracranial pressure
25:763-774. monitoring technology. J Neurotrauma 2007, 24 Suppl 1:S45-S54.
3. Pellerin L, Bouzier-Sore AK, Aubert A, Serres S, Merle M, Costalat R, Magistretti 23. Vajkoczy P, Roth H, Horn P, Lucke T, Thome C, Hubner U, Martin GT, Zappletal
PJ: Activity-dependent regulation of energy metabolism by astrocytes: C, Klar E, Schilling L, Schmiedek P: Continuous monitoring of regional
an update. Glia 2007, 55:1251-1262. cerebral blood flow: experimental and clinical validation of a novel
4. Bouzier-Sore AK, Voisin P, Canioni P, Magistretti PJ, Pellerin L: Lactate is a thermal diffusion microprobe. J Neurosurg 2000, 93:265-274.
preferential oxidative energy substrate over glucose for neurons in 24. Muench E, Horn P, Bauhuf C, Roth H, Philipps M, Hermann P, Quintel M,
culture. J Cereb Blood Flow Metab 2003, 23:1298-1306. Schmiedek P, Vajkoczy P: Effects of hypervolemia and hypertension on
5. Oddo M, Levine JM, Frangos S, Maloney-Wilensky E, Carrera E, Daniel RT, regional cerebral blood flow, intracranial pressure, and brain tissue
Levivier M, Magistretti PJ, LeRoux PD: Brain lactate metabolism in humans oxygenation after subarachnoid hemorrhage. Crit Care Med 2007,
with subarachnoid hemorrhage. Stroke 2012, 43:1418-1421. 35:1844-1851.
6. Coles JP, Fryer TD, Smielewski P, Chatfield DA, Steiner LA, Johnston AJ, 25. Rosenthal G, Sanchez-Mejia RO, Phan N, Hemphill JC, III, Martin C, Manley GT:
Downey SP, Williams GB, Aigbirhio F, Hutchinson PJ, Rice K, Carpenter TA, Clark Incorporating a parenchymal thermal diffusion cerebral blood flow probe
JC, Pickard JD, Menon DK: Incidence and mechanisms of cerebral ischemia in bedside assessment of cerebral autoregulation and vasoreactivity in
in early clinical head injury. J Cereb Blood Flow Metab 2004, 24:202-211. patients with severe traumatic brain injury. J Neurosurg 2011, 114:62-70.
7. Czosnyka M, Smielewski P, Piechnik S, Steiner LA, Pickard JD: Cerebral 26. Washington CW, Zipfel GJ: Detection and monitoring of vasospasm and
autoregulation following head injury. J Neurosurg 2001, 95:756-763. delayed cerebral ischemia: a review and assessment of the literature.
8. Coles JP, Steiner LA, Johnston AJ, Fryer TD, Coleman MR, Smieleweski P, Neurocrit Care 2011, 15:312-317.
Chatfield DA, Aigbirhio F, Williams GB, Boniface S, Rice K, Clark JC, Pickard JD, 27. de Riva N., Budohoski KP, Smielewski P, Kasprowicz M, Zweifel C, Steiner LA,
Menon DK: Does induced hypertension reduce cerebral ischaemia within Reinhard M, Fabregas N, Pickard JD, Czosnyka M: Transcranial Doppler
the traumatized human brain? Brain 2004, 127:2479-2490. Pulsatility Index: what it is and what it isn’t. Neurocrit Care 2012, 17:58-66.
9. Rosenthal G, Hemphill JC, III, Sorani M, Martin C, Morabito D, Obrist WD, 28. Steiner LA, Coles JP, Johnston AJ, Chatfield DA, Smielewski P, Fryer TD,
Manley GT: Brain tissue oxygen tension is more indicative of oxygen Aigbirhio FI, Clark JC, Pickard JD, Menon DK, Czosnyka M: Assessment of
diffusion than oxygen delivery and metabolism in patients with traumatic cerebrovascular autoregulation in head-injured patients: a validation
brain injury. Crit Care Med 2008, 36:1917-1924. study. Stroke 2003, 34:2404-2409.
10. Menon DK, Coles JP, Gupta AK, Fryer TD, Smielewski P, Chatfield DA, Aigbirhio 29. Chieregato A, Sabia G, Tanfani A, Compagnone C, Tagliaferri F, Targa L: Xenon-
F, Skepper JN, Minhas PS, Hutchinson PJ, Carpenter TA, Clark JC, Pickard JD: CT and transcranial Doppler in poor-grade or complicated aneurysmatic
Diffusion limited oxygen delivery following head injury. Crit Care Med 2004, subarachnoid hemorrhage patients undergoing aggressive management
32:1384-1390. of intracranial hypertension. Intensive Care Med 2006, 32:1143-1150.
11. Teasdale G, Jennett B: Assessment of coma and impaired consciousness. 30. Zonta M, Angulo MC, Gobbo S, Rosengarten B, Hossmann KA, Pozzan T,
A practical scale. Lancet 1974, 2:81-84. Carmignoto G: Neuron-to-astrocyte signaling is central to the dynamic
12. Wijdicks EF, Bamlet WR, Maramattom BV, Manno EM, McClelland RL: control of brain microcirculation. Nat Neurosci 2003, 6:43-50.
Validation of a new coma scale: The FOUR score. Ann Neurol 2005, 31. Panerai RB: Assessment of cerebral pressure autoregulation in humans--a
58:585-593. review of measurement methods. Physiol Meas 1998, 19:305-338.
13. Ely EW, Margolin R, Francis J, May L, Truman B, Dittus R, Speroff T, Gautam S, 32. De Backer D, Ospina-Tascon G, Salgado D, Favory R, Creteur J, Vincent JL:
Bernard GR, Inouye SK: Evaluation of delirium in critically ill patients: Monitoring the microcirculation in the critically ill patient: current
validation of the Confusion Assessment Method for the Intensive Care methods and future approaches. Intensive Care Med 2010, 36:1813-1825.
Unit (CAM-ICU). Crit Care Med 2001, 29:1370-1379. 33. De Backer D, Donadello K, Taccone FS, Ospina-Tascon G, Salgado D, Vincent
14. Bergeron N, Dubois MJ, Dumont M, Dial S, Skrobik Y: Intensive Care Delirium JL: Microcirculatory alterations: potential mechanisms and implications for
Screening Checklist: evaluation of a new screening tool. Intensive Care Med therapy. Ann Intensive Care 2011, 1:27.
2001, 27:859-864. 34. Perez-Barcena J, Goedhart P, Ibanez J, Brell M, Garcia R, Llinas P, Jimenez C,
15. Marmarou A, Anderson RL, Ward JD, Choi SC, Young HF, Eisenberg HM, Ince C: Direct observation of human microcirculation during
Foulkes MA, Marshall LF, Jane JA: Impact of ICP instability and hypotension decompressive craniectomy after stroke. Crit Care Med 2011, 39:1126-1129.
on outcome in patients with severe head trauma. J Neurosurg Special Suppl 35. Pennings FA, Albrecht KW, Muizelaar JP, Schuurman PR, Bouma GJ: Abnormal
1991, 75:S59-S66. responses of the human cerebral microcirculation to papaverin during
16. Bratton SL, Chestnut RM, Ghajar J, McConnell Hammond FF, Harris OA, Hartl aneurysm surgery. Stroke 2009, 40:317-320.
R, Manley GT, Nemecek A, Newell DW, Rosenthal G, Schouten J, Shutter L, 36. Chieregato A, Calzolari F, Trasforini G, Targa L, Latronico N: Normal jugular
Timmons SD, Ullman JS, Videtta W, Wilberger JE, Wright DW: Guidelines for bulb oxygen saturation. J Neurol Neurosurg Psychiatry 2003, 74:784-786.
the management of severe traumatic brain injury. VI. Indications for 37. Verweij BH, Muizelaar JP, Vinas FC: Hyperacute measurement of intracranial
intracranial pressure monitoring. J Neurotrauma 2007, 24 Suppl 1:S37-S44. pressure, cerebral perfusion pressure, jugular venous oxygen saturation,
17. Bederson JB, Connolly ES Jr, Batjer HH, Dacey RG, Dion JE, Diringer MN, and laser Doppler flowmetry, before and during removal of traumatic
Duldner JE Jr, Harbaugh RE, Patel AB, Rosenwasser RH: Guidelines for the acute subdural hematoma. J Neurosurg 2001, 95:569-572.
management of aneurysmal subarachnoid hemorrhage: a statement for 38. Gopinath SP, Robertson CS, Contant CF, Hayes C, Feldman Z, Narayan RK,
healthcare professionals from a special writing group of the Stroke Grossman RG: Jugular venous desaturation and outcome after head injury.
Council, American Heart Association. Stroke 2009, 40:994-1025. J Neurol Neurosurg Psychiatry 1994, 57:717-723.
18. Morgenstern LB, Hemphill JC, III, Anderson C, Becker K, Broderick JP, Connolly 39. Shenkin HA, Harmel MH, Kety SS: Dynamic anatomy of the cerebral
ES Jr, Greenberg SM, Huang JN, MacDonald RL, Messe SR, Mitchell PH, Selim circulation. Arch Neurol Psychiatry 1948, 60:240-252.
M, Tamargo RJ: Guidelines for the management of spontaneous 40. Coles JP, Fryer TD, Coleman MR, Smielewski P, Gupta AK, Minhas PS, Aigbirhio
intracerebral hemorrhage: a guideline for healthcare professionals from F, Chatfield DA, Williams GB, Boniface S, Carpenter TA, Clark JC, Pickard JD,
the American Heart Association/American Stroke Association. Stroke 2010, Menon DK: Hyperventilation following head injury: effect on ischemic
41:2108-2129. burden and cerebral oxidative metabolism. Crit Care Med 2007, 35:568-578.
19. Pickard JD, Czosnyka M: Management of raised intracranial pressure. 41. Stocchetti N, Paparella A, Bridelli F, Bacchi M, Piazza P, Zuccoli P: Cerebral
J Neurol Neurosurg Psychiatry 1993, 56:845-858. venous oxygen saturation studied with bilateral samples in the internal
20. Bouzat P, Francony G, Fauvage B, Payen JF: Transcranial Doppler pulsatility jugular veins. Neurosurgery 1994, 34:38-43.
index for initial management of brain-injured patients. Neurosurgery 2010, 42. Ponce LL, Pillai S, Cruz J, Li X, Julia H, Gopinath S, Robertson CS: Position of
67:E1863-E1864. probe determines prognostic information of brain tissue PO2 in severe
21. Rajajee V, Vanaman M, Fletcher JJ, Jacobs TL: Optic nerve ultrasound for the traumatic brain injury. Neurosurgery 2012, 70:1492-1503.
detection of raised intracranial pressure. Neurocrit Care 2011, 15:506-515. 43. Scheufler KM, Lehnert A, Rohrborn HJ, Nadstawek J, Thees C: Individual value
22. Bratton SL, Chestnut RM, Ghajar J, McConnell Hammond FF, Harris OA, Hartl of brain tissue oxygen pressure, microvascular oxygen saturation,
Stocchetti et al. Critical Care 2013, 17:201 Page 12 of 13
http://ccforum.com/content/17/1/201

cytochrome redox level, and energy metabolites in detecting critically panel method for outcome prediction following aneurysmal
reduced cerebral energy state during acute changes in global cerebral subarachnoid hemorrhage. Intensive Care Med 2010, 36:107-115.
perfusion. J Neurosurg Anesthesiol 2004, 16:210-219. 63. Vos PE, Lamers KJ, Hendriks JC, van Haaren M, Beems T, Zimmerman C,
44. Chang JJ, Youn TS, Benson D, Mattick H, Andrade N, Harper CR, Moore CB, van Geel W, de Reus H, Biert J, Verbeek MM: Glial and neuronal proteins in
Madden CJ, Diaz-Arrastia RR: Physiologic and functional outcome correlates serum predict outcome after severe traumatic brain injury. Neurology 2004,
of brain tissue hypoxia in traumatic brain injury. Crit Care Med 2009, 62:1303-1310.
37:283-290. 64. Korfias S, Stranjalis G, Boviatsis E, Psachoulia C, Jullien G, Gregson B,
45. Hlatky R, Valadka AB, Goodman JC, Contant CF, Robertson CS: Patterns of Mendelow AD, Sakas DE: Serum S-100B protein monitoring in patients with
energy substrates during ischemia measured in the brain by microdialysis. severe traumatic brain injury. Intensive Care Med 2007, 33:255-260.
J Neurotrauma 2004, 21:894-906. 65. Zandbergen EG, Hijdra A, Koelman JH, Hart AA, Vos PE, Verbeek MM, de Haan
46. Dings J, Meixensberger J, Jager A, Roosen K: Clinical experience with 118 RJ: Prediction of poor outcome within the first 3 days of postanoxic coma.
brain tissue oxygen partial pressure catheter probes. Neurosurgery 1998, Neurology 2006, 66:62-68.
43:1082-1095. 66. Sanchez-Pena P, Pereira AR, Sourour NA, Biondi A, Lejean L, Colonne C, Boch
47. van den Brink WA, van Santbrink H, Steyerberg EW, Avezaat CJ, Suazo JA, AL, Al Hawari M, Abdennour L, Puybasset L: S100B as an additional
Hogesteeger C, Jansen WJ, Kloos LM, Vermeulen J, Maas AI: Brain oxygen prognostic marker in subarachnoid aneurysmal hemorrhage. Crit Care Med
tension in severe head injury. Neurosurgery 2000, 46:868-876. 2008, 36:2267-2273.
48. Oddo M, Levine JM, Mackenzie L, Frangos S, Feihl F, Kasner SE, Katsnelson M, 67. Sen J, Belli A: S100B in neuropathologic states: the CRP of the brain?
Pukenas B, MacMurtrie E, Maloney-Wilensky E, Kofke WA, LeRoux PD: Brain J Neurosci Res 2007, 85:1373-1380.
hypoxia is associated with short-term outcome after severe traumatic 68. Vos PE, Jacobs B, Andriessen TM, Lamers KJ, Borm GF, Beems T, Edwards M,
brain injury independently of intracranial hypertension and low cerebral Rosmalen CF, Vissers JL: GFAP and S100B are biomarkers of traumatic brain
perfusion pressure. Neurosurgery 2011, 69:1037-1045. injury: an observational cohort study. Neurology 2010, 75:1786-1793.
49. Longhi L, Valeriani V, Rossi S, De Marchi M, Egidi M, Stocchetti N: Effects of 69. Herrmann M, Vos P, Wunderlich MT, de Bruijn CH, Lamers KJ: Release of glial
hyperoxia on brain tissue oxygen tension in cerebral focal lesions. Acta tissue-specific proteins after acute stroke: A comparative analysis of
Neurochir Suppl 2002, 81:315-317. serum concentrations of protein S-100B and glial fibrillary acidic protein.
50. Nangunoori R, Maloney-Wilensky E, Stiefel M, Park S, Andrew KW, Levine JM, Stroke 2000, 31:2670-2677.
Yang W, Le Roux PD: Brain tissue oxygen-based therapy and outcome after 70. Vos PE, van Gils M, Beems T, Zimmerman C, Verbeek MM: Increased GFAP
severe traumatic brain injury: A systematic literature review. Neurocrit Care and S100beta but not NSE serum levels after subarachnoid haemorrhage
2012, 17:131-138. are associated with clinical severity. Eur J Neurol 2006, 13:632-638.
51. Leal-Noval SR, Cayuela A, rellano-Orden V, Marin-Caballos A, Padilla V, 71. Kaneko T, Kasaoka S, Miyauchi T, Fujita M, Oda Y, Tsuruta R, Maekawa T: Serum
Ferrandiz-Millon C, Corcia Y, Garcia-Alfaro C, maya-Villar R, Murillo-Cabezas F: glial fibrillary acidic protein as a predictive biomarker of neurological
Invasive and noninvasive assessment of cerebral oxygenation in patients outcome after cardiac arrest. Resuscitation 2009, 80:790-794.
with severe traumatic brain injury. Intensive Care Med 2010, 36:1309-1317. 72. Pelinka LE, Kroepfl A, Leixnering M, Buchinger W, Raabe A, Redl H: GFAP
52. Naidech AM, Bendok BR, Ault ML, Bleck TP: Monitoring with the Somanetics versus S100B in serum after traumatic brain injury: relationship to brain
INVOS 5100C after aneurysmal subarachnoid hemorrhage. Neurocrit Care damage and outcome. J Neurotrauma 2004, 21:1553-1561.
2008, 9:326-331. 73. Cronberg T, Rundgren M, Westhall E, Englund E, Siemund R, Rosen I, Widner
53. Hillered L, Vespa PM, Hovda DA: Translational neurochemical research in H, Friberg H: Neuron-specific enolase correlates with other prognostic
acute human brain injury: the current status and potential future for markers after cardiac arrest. Neurology 2011, 77:623-630.
cerebral microdialysis. J Neurotrauma 2005, 22:3-41. 74. Ost M, Nylen K, Csajbok L, Ohrfelt AO, Tullberg M, Wikkelso C, Nellgard P,
54. Andrews PJ, Citerio G, Longhi L, Polderman K, Sahuquillo J, Vajkoczy P: NICEM Rosengren L, Blennow K, Nellgard B: Initial CSF total tau correlates with
consensus on neurological monitoring in acute neurological disease. 1-year outcome in patients with traumatic brain injury. Neurology 2006,
Intensive Care Med 2008, 34:1362-1370. 67:1600-1604.
55. Bellander BM, Cantais E, Enblad P, Hutchinson P, Nordstrom CH, Robertson C, 75. Magnoni S, Esparza TJ, Conte V, Carbonara M, Carrabba G, Holtzman DM,
Sahuquillo J, Smith M, Stocchetti N, Ungerstedt U, Unterberg A, Olsen NV: Zipfel GJ, Stocchetti N, Brody DL: Tau elevations in the brain extracellular
Consensus meeting on microdialysis in neurointensive care. Intensive Care space correlate with reduced amyloid-beta levels and predict adverse
Med 2004, 30:2166-2169. clinical outcomes after severe traumatic brain injury. Brain 2012,
56. Sarrafzadeh A, Haux D, Sakowitz O, Benndorf G, Herzog H, Kuechler I, 135:1268-1280.
Unterberg A: Acute focal neurological deficits in aneurysmal subarachnoid 76. Kay A, Petzold A, Kerr M, Keir G, Thompson E, Nicoll J: Temporal alterations in
hemorrhage: relation of clinical course, CT findings, and metabolite cerebrospinal fluid amyloid beta-protein and apolipoprotein E after
abnormalities monitored with bedside microdialysis. Stroke 2003, subarachnoid hemorrhage. Stroke 2003, 34:e240-e243.
34:1382-1388. 77. Franz G, Beer R, Kampfl A, Engelhardt K, Schmutzhard E, Ulmer H,
57. Oddo M, Schmidt JM, Carrera E, Badjatia N, Connolly ES, Presciutti M, Deisenhammer F: Amyloid beta 1-42 and tau in cerebrospinal fluid after
Ostapkovich ND, Levine JM, Le Roux P, Mayer SA: Impact of tight glycemic severe traumatic brain injury. Neurology 2003, 60:1457-1461.
control on cerebral glucose metabolism after severe brain injury: a 78. Brody DL, Magnoni S, Schwetye KE, Spinner ML, Esparza TJ, Stocchetti N,
microdialysis study. Crit Care Med 2008, 36:3233-3238. Zipfel GJ, Holtzman DM: Amyloid-beta dynamics correlate with
58. Ko SB, Choi HA, Parikh G, Helbok R, Schmidt JM, Lee K, Badjatia N, Claassen J, neurological status in the injured human brain. Science 2008,
Connolly ES, Mayer SA: Multimodality monitoring for cerebral perfusion 321:1221-1224.
pressure optimization in comatose patients with intracerebral 79. Petzold A, Tisdall MM, Girbes AR, Martinian L, Thom M, Kitchen N, Smith M:
hemorrhage. Stroke 2011, 42:3087-3092. In vivo monitoring of neuronal loss in traumatic brain injury:
59. Schmidt JM, Ko SB, Helbok R, Kurtz P, Stuart RM, Presciutti M, Fernandez L, Lee a microdialysis study. Brain 2011, 134:464-483.
K, Badjatia N, Connolly ES, Claassen J, Mayer SA: Cerebral perfusion pressure 80. Petzold A, Keir G, Kay A, Kerr M, Thompson EJ: Axonal damage and outcome
thresholds for brain tissue hypoxia and metabolic crisis after poor-grade in subarachnoid haemorrhage. J Neurol Neurosurg Psychiatry 2006,
subarachnoid hemorrhage. Stroke 2011, 42:1351-1356. 77:753-759.
60. Oddo M, Milby A, Chen I, Frangos S, MacMurtrie E, Maloney-Wilensky E, Stiefel 81. Petzold A, Rejdak K, Belli A, Sen J, Keir G, Kitchen N, Smith M, Thompson EJ:
M, Kofke WA, Levine JM, Le Roux PD: Hemoglobin concentration and Axonal pathology in subarachnoid and intracerebral hemorrhage.
cerebral metabolism in patients with aneurysmal subarachnoid J Neurotrauma 2005, 22:407-414.
hemorrhage. Stroke 2009, 40:1275-1281. 82. Papa L, Akinyi L, Liu MC, Pineda JA, Tepas JJ, III, Oli MW, Zheng W, Robinson G,
61. Laskowitz DT, Kasner SE, Saver J, Remmel KS, Jauch EC: Clinical usefulness of Robicsek SA, Gabrielli A, Heaton SC, Hannay HJ, Demery JA, Brophy GM,
a biomarker-based diagnostic test for acute stroke: the Biomarker Rapid Layon J, Robertson CS, Hayes RL, Wang KK: Ubiquitin C-terminal hydrolase is
Assessment in Ischemic Injury (BRAIN) study. Stroke 2009, 40:77-85. a novel biomarker in humans for severe traumatic brain injury. Crit Care
62. Turck N, Vutskits L, Sanchez-Pena P, Robin X, Hainard A, Gex-Fabry M, Fouda C, Med 2010, 38:138-144.
Bassem H, Mueller M, Lisacek F, Puybasset L, Sanchez JC: A multiparameter 83. Mondello S, Papa L, Buki A, Bullock MR, Czeiter E, Tortella FC, Wang KK, Hayes
Stocchetti et al. Critical Care 2013, 17:201 Page 13 of 13
http://ccforum.com/content/17/1/201

RL: Neuronal and glial markers are differently associated with computed 99. Drenckhahn C, Winkler MK, Major S, Scheel M, Kang EJ, Pinczolits A, Grozea C,
tomography findings and outcome in patients with severe traumatic brain Hartings JA, Woitzik J, Dreier JP: Correlates of spreading depolarization in
injury: a case control study. Crit Care 2011, 15:R156. human scalp electroencephalography. Brain 2012, 135:853-868.
84. Pineda JA, Lewis SB, Valadka AB, Papa L, Hannay HJ, Heaton SC, Demery JA, 100. Robinson LR, Micklesen PJ, Tirschwell DL, Lew HL: Predictive value of
Liu MC, Aikman JM, Akle V, Brophy GM, Tepas JJ, Wang KK, Robertson CS, somatosensory evoked potentials for awakening from coma. Crit Care Med
Hayes RL: Clinical significance of alphaII-spectrin breakdown products in 2003, 31:960-967.
cerebrospinal fluid after severe traumatic brain injury. J Neurotrauma 2007, 101. Oddo M, Rossetti AO: Predicting neurological outcome after cardiac arrest.
24:354-366. Curr Opin Crit Care 2011, 17:254-259.
85. Berger RP, Hayes RL, Richichi R, Beers SR, Wang KK: Serum concentrations of 102. Hakimi K, Kinney G, Kraft G, Micklesen P, Robinson L: Reliability in
ubiquitin C-terminal hydrolase-L1 and alphaII-spectrin breakdown interpretation of median somatosensory evoked potentials in the setting
product 145 kDa correlate with outcome after pediatric TBI. J Neurotrauma of coma: factors and implications. Neurocrit Care 2009, 11:353-361.
2012, 29:162-167. 103. Zandbergen EG, Koelman JH, de Haan RJ, Hijdra A: SSEPs and prognosis in
86. Vespa PM, Miller C, McArthur D, Eliseo M, Etchepare M, Hirt D, Glenn TC, postanoxic coma: only short or also long latency responses? Neurology
Martin N, Hovda D: Nonconvulsive electrographic seizures after traumatic 2006, 67:583-586.
brain injury result in a delayed, prolonged increase in intracranial pressure 104. Marshall LF, Marshall SB, Klauber MR, van Berkum Clark M, Eisenberg HM, Jane
and metabolic crisis. Crit Care Med 2007, 35:2830-2836. JA, Luerssen TG, Marmarou A, Foulkes MA: A new classification of head
87. Pandian JD, Cascino GD, So EL, Manno E, Fulgham JR: Digital video- injury based on computerized tomography. J Neurosurg 1991, 75:S14-S20.
electroencephalographic monitoring in the neurological-neurosurgical 105. Maas AI, Hukkelhoven CW, Marshall LF, Steyerberg EW: Prediction of
intensive care unit: clinical features and outcome. Arch Neurol 2004, outcome in traumatic brain injury with computed tomographic
61:1090-1094. characteristics: a comparison between the computed tomographic
88. Claassen J, Mayer SA, Kowalski RG, Emerson RG, Hirsch LJ: Detection of classification and combinations of computed tomographic predictors.
electrographic seizures with continuous EEG monitoring in critically ill Neurosurgery 2005, 57:1173-1182.
patients. Neurology 2004, 62:1743-1748. 106. Wijman CA, Mlynash M, Caulfield AF, Hsia AW, Eyngorn I, Bammer R, Fischbein
89. Vespa PM, Nuwer MR, Juhasz C, Alexander M, Nenov V, Martin N, Becker DP: N, Albers GW, Moseley M: Prognostic value of brain diffusion-weighted
Early detection of vasospasm after acute subarachnoid hemorrhage using imaging after cardiac arrest. Ann Neurol 2009, 65:394-402.
continuous EEG ICU monitoring. Electroencephalogr Clin Neurophysiol 1997, 107. Sidaros A, Engberg AW, Sidaros K, Liptrot MG, Herning M, Petersen P, Paulson
103:607-615. OB, Jernigan TL, Rostrup E: Diffusion tensor imaging during recovery from
90. Rathakrishnan R, Gotman J, Dubeau F, Angle M: Using continuous severe traumatic brain injury and relation to clinical outcome:
electroencephalography in the management of delayed cerebral ischemia a longitudinal study. Brain 2008, 131:559-572.
following subarachnoid hemorrhage. Neurocrit Care 2011, 14:152-161. 108. Tollard E, Galanaud D, Perlbarg V, Sanchez-Pena P, Le Fur Y, Abdennour L,
91. Klopman MA, Sebel PS: Cost-effectiveness of bispectral index monitoring. Cozzone P, Lehericy S, Chiras J, Puybasset L: Experience of diffusion tensor
Curr Opin Anaesthesiol 2011, 24:177-181. imaging and 1H spectroscopy for outcome prediction in severe traumatic
92. Boccagni C, Bagnato S, Sant AA, Prestandrea C, Galardi G: Usefulness of brain injury: Preliminary results. Crit Care Med 2009, 37:1448-1455.
standard EEG in predicting the outcome of patients with disorders of 109. Tong KA, Ashwal S, Holshouser BA, Nickerson JP, Wall CJ, Shutter LA,
consciousness after anoxic coma. J Clin Neurophysiol 2011, 28:489-492. Osterdock RJ, Haacke EM, Kido D: Diffuse axonal injury in children: clinical
93. Vespa PM, Boscardin WJ, Hovda DA, McArthur DL, Nuwer MR, Martin NA, correlation with hemorrhagic lesions. Ann Neurol 2004, 56:36-50.
Nenov V, Glenn TC, Bergsneider M, Kelly DF, Becker DP: Early and persistent 110. Kasahara M, Menon DK, Salmond CH, Outtrim JG, Taylor Tavares JV, Carpenter
impaired percent alpha variability on continuous electroencephalography TA, Pickard JD, Sahakian BJ, Stamatakis EA: Altered functional connectivity in
monitoring as predictive of poor outcome after traumatic brain injury. the motor network after traumatic brain injury. Neurology 2010,
J Neurosurg 2002, 97:84-92. 75:168-176.
94. Hebb MO, McArthur DL, Alger J, Etchepare M, Glenn TC, Bergsneider M, 111. Karakis I, Montouris GD, Otis JA, Douglass LM, Jonas R, Velez-Ruiz N, Wilford K,
Martin N, Vespa PM: Impaired percent alpha variability on continuous Espinosa PS: A quick and reliable EEG montage for the detection of
electroencephalography is associated with thalamic injury and predicts seizures in the critical care setting. J Clin Neurophysiol 2010, 27:100-105.
poor long-term outcome after human traumatic brain injury. 112. Piper I, Chambers I, Citerio G, Enblad P, Gregson B, Howells T, Kiening K,
J Neurotrauma 2007, 24:579-590. Mattern J, Nilsson P, Ragauskas A, Sahuquillo J, Donald R, Sinnott R, Stell A:
95. Dreier JP: The role of spreading depression, spreading depolarization and The brain monitoring with Information Technology (BrainIT) collaborative
spreading ischemia in neurological disease. Nat Med 2011, 17:439-447. network: EC feasibility study results and future direction. Acta Neurochir
96. Feuerstein D, Manning A, Hashemi P, Bhatia R, Fabricius M, Tolias C, Pahl C, (Wien ) 2010, 152:1859-1871.
Ervine M, Strong AJ, Boutelle MG: Dynamic metabolic response to multiple 113. Sorani MD, Hemphill JC, III, Morabito D, Rosenthal G, Manley GT: New
spreading depolarizations in patients with acute brain injury: an online approaches to physiological informatics in neurocritical care. Neurocrit
microdialysis study. J Cereb Blood Flow Metab 2010, 30:1343-1355. Care 2007, 7:45-52.
97. Hartings JA, Watanabe T, Bullock MR, Okonkwo DO, Fabricius M, Woitzik J, 114. Maas AI, Menon DK, Lingsma HF, Pineda JA, Sandel ME, Manley GT: Re-
Dreier JP, Puccio A, Shutter LA, Pahl C, Strong AJ: Spreading depolarizations orientation of clinical research in traumatic brain injury: report of an
have prolonged direct current shifts and are associated with poor international workshop on comparative effectiveness research.
outcome in brain trauma. Brain 2011, 134:1529-1540. J Neurotrauma 2012, 29:32-46.
98. Woitzik J, Dreier JP, Hecht N, Fiss I, Sandow N, Major S, Winkler M, Dahlem YA,
Manville J, Diepers M, Muench E, Kasuya H, Schmiedek P, Vajkoczy P: Delayed
doi:10.1186/cc11513
cerebral ischemia and spreading depolarization in absence of
Cite this article as: Stocchetti N, et al.: Clinical review: Neuromonitoring -
angiographic vasospasm after subarachnoid hemorrhage. J Cereb Blood an update. Critical Care 2013, 17:201.
Flow Metab 2012, 32:203-212.

You might also like