You are on page 1of 15

Neurocrit Care

https://doi.org/10.1007/s12028-019-00814-4

INTRACEREBRAL HEMORRHAGE

Emergency Neurological Life Support:


Intracerebral Hemorrhage
Arthur M. Lam1*, Vineeta Singh2 and A. M. Iqbal O’Meara3

© 2019 Neurocritical Care Society

Abstract 
Intracerebral hemorrhage (ICH) is a subset of stroke due to spontaneous bleeding within the parenchyma of the
brain. It is potentially lethal, and survival depends on ensuring an adequate airway, proper diagnosis, and early man-
agement of several specific issues such as blood pressure, coagulopathy reversal, and surgical hematoma evacuation
for appropriate patients. ICH was chosen as an Emergency Neurological Life Support protocol because intervention
within the first hours may improve outcome, and it is critical to have site-specific protocols to drive care quickly and
efficiently. This module is meant to give a broad framework for the principles of diagnosis and emergent manage-
ment of ICH, which can be adapted to reflect global and regional variations based on the local availability of diagnos-
tic tools and treatments such as a specific agent for reversal of coagulopathy or for management of hypertension.
Keywords:  Intracerebral hemorrhage, Blood pressure, Hematoma, Coagulopathy, Surgery, External ventricular drain

Introduction ICH patient is recommended, in accordance with exist-


Intracerebral hemorrhage (ICH) results from sponta- ing guidelines [1, 2].
neous direct bleeding into the brain. In the USA, ICH Management of the ICH patient during the initial
accounts for 10–15% of all strokes, but it carries a dispro- “golden hour” emphasizes the following aspects:
portionately high risk of death or long-term disability. It
is considered an acute neurological emergency because 1. Stabilization and frequent reassessment of the
of the potential to treat or mitigate injury from the ICH patient’s airway, breathing, and circulation (ABCs).
and attenuate the risk of ongoing secondary brain injury. 2. Rapid and accurate diagnosis using neuroimaging.
The availability of treatments proven to benefit ICH 3. Concise clinical assessment regarding ICH charac-
patients has lagged behind that of ischemic stroke and teristics and patient condition.
aneurysmal subarachnoid hemorrhage, and this has 4. Targeted assessment for potential early interventions
resulted in variability in care that ranges from aggressive including:
treatment to a nihilistic approach. Guidelines exist for
the management of ICH, and the purpose of this Emer- a. Control of elevated blood pressure.
gency Neurological Life Support (ENLS) protocol is to b. Correction of coagulopathy.
emphasize initial management, with the goal of optimiz- c. Need for early surgical intervention.
ing recovery. Acknowledging that there is variability in 5. Anticipation of specific patient care needs such as:
the strength of evidence for treatment recommendations
for certain interventions, aggressive initial care of the a. Specific treatment aspects related to underlying
ICH cause.
b. Risk of early clinical deterioration and hema-
*Correspondence: aml150@ucsd.edu
1
toma expansion.
Department of Anesthesiology, University of California, 200W Arbor St,
San Diego, CA 94110, USA
c. Need for intracranial pressure (ICP) or other
Full list of author information is available at the end of the article neuromonitoring.
d. Patient disposition from the emergency depart-
ment (ED).
Management Protocol Table 1  ICH checklist for the first hour
The ENLS-suggested algorithm for the initial manage- Checklist
ment of ICH is shown in Fig. 1. Suggested items to com-
plete within the first hour of evaluating a patient with ICH ☐ Complete blood count with platelet count, PT, PTT, INR
are shown in Table 1. After initial management of airway, ☐ CT head results: hematoma size, location, presence of intraventricular
hemorrhage
breathing, and circulation, and the diagnosis of ICH is
☐ Glasgow Coma Scale (GCS) score
made, all five steps should be addressed simultaneously
☐ Calculate ICH score
(history, BP control, hemostasis, surgery, and EVD) with
☐ Interventions:
specific management dictated by the patient’s condition.    ☐ Coagulopathy reversal (goal INR ≤ 1.4)
Transfer to a neuro-ICU which has physician and nurs-    ☐ Blood pressure lowering (goal systolic 140–180 mmHg)
ing expertise in management of ICH patients is recom-    ☐ Surgical hematoma evacuation (if indicated)
   ☐ Airway/ventilation management
mended and is associated with improved outcomes.

Prehospital Considerations
The initial prehospital and ED resuscitation is similar acute ischemic stroke are different, ICH-specific inter-
across stroke subtypes, with rapid neuroimaging being ventions are not provided until the diagnosis is made.
essential to diagnosis. Because treatments for ICH and Thus, prehospital care focuses on management of the

Fig. 1  ENLS ICH management algorithm


ABCs, treatment of rapidly reversible causes of neuro- logistics related to availability and the clinical condition
logical deficit (e.g., hypoglycemia), early radio notifica- of the patient limit its use as a primary modality [3, 4].
tion, and rapid transport to a designated stroke receiving
hospital.
As with all emergency medical care, the initial assess- Interpreting the CT Scan: Location, Volume, Spot Sign,
ment of the ABCs is critical. Obtunded or comatose and the BAT Score
patients will need intubation for airway protection and/ ICH tends to occur in characteristic locations, with
or adequate ventilation aimed at normocapnia. Fluid hypertensive ICH most frequently located in the basal
infusion and volume replacement to maintain an ade- ganglia, thalamus, pons (brainstem), and cerebellum.
quate blood pressure should be initiated as needed. As ICH due to cerebral amyloid angiopathy or AVM tends
with all strokes, it is important to obtain a brief history to have a lobar location. The origin of the hematoma is
on the onset of symptoms and when the patient was last usually evident from the initial CT scan, and its location
seen normal. Prehospital providers should attempt to get influences outcome and treatment (Fig. 2).
a medical history and list of medications prescribed for While ICH location is important, ICH hematoma vol-
the patient with specific attention to antiplatelets, anti- ume is a stronger predictor of patient outcome. The abil-
coagulants, and antihypertensive medications. Triage to ity to calculate hematoma volume quickly from the initial
an appropriate emergency facility for stroke management CT scan is an advantage in directing communication and
should be done promptly and expeditiously. Suggested treatment decisions. Automated CT software algorithms
sample “hand-off ” language to the ED team by paramed- can be used to calculate hematoma volume. However,
ics is included in Table 4. the manual ABC/2 formula, which approximates the vol-
ume of an ellipsoid, is simple and reasonably accurate
Diagnosis compared to computerized methods [5]. When using
ICH may result from a variety of underlying etiologies. the ABC/2 method for calculating volume, the axial CT
Rupture of a small arteriole(s) due to chronic hyperten- image is selected with the largest cross-sectional area of
sion accounts for approximately 60% of cases. Other hemorrhage. Figure 3 demonstrates an example of calcu-
common causes include cerebral amyloid angiopathy, lating ICH volume by ABC/2 method.
coagulopathy due to treatment with antithrombotic and Many ICH patients (up to 40%) experience hematoma
anticoagulant medications, sympathomimetic drugs such growth after initial presentation, and the ability to antici-
as cocaine, and underlying vascular anomalies such as pate expansion is desirable, as expansion is associated
arteriovenous malformations (AVMs) or cavernous mal- with worse clinical outcome [6]. Several retrospective
formations. Less common causes include cerebral vascu- reports have suggested that the use of intravenous (IV)
litis, Moya–Moya syndrome, and rupture of a saccular or contrast administration during the initial CT scan may
mycotic aneurysm. Secondary hemorrhagic transforma- identify extravasation into the hematoma and that this
tion of an arterial or venous infarct may also occur. “spot sign” (contrast within the hematoma) is predictive
Most patients with acute ICH develop the sudden onset of hematoma growth (Fig. 4) [7–9].
of a focal neurological abnormality. Without neuroimag- Thus, the use of a “stroke CT” that includes non-con-
ing, ICH often cannot be reliably distinguished from an trast CT as well as CT angiography (and possibly CT
acute ischemic stroke. Headache, progressive neurologi- perfusion and post-contrast images) may be consid-
cal signs and symptoms, acute severe hypertension, and ered in patients presenting with acute stroke (ischemic
decreased level of consciousness occur more frequently or ICH). In acute ICH, this can detect a “spot sign,” as
in ICH than in ischemic stroke. well as reveal an underlying vascular anomaly. Ongo-
ing studies are seeking to use the “spot sign” as a way to
Neuroimaging identify those at risk of hemorrhage expansion and to
Non-contrast computed tomography (CT) is the most determine whether hemostatic agents may benefit these
commonly used modality given that it can be done specific high-risk patients. However, CT angiography is
quickly, can be used for critically ill patients, and has a not performed in many centers, and recently, the BAT
very high sensitivity and specificity for acute parenchy- score based on non-contrast CT has been reported to
mal hemorrhage. Magnetic resonance imaging (MRI) have a 50% sensitivity, 89% specificity, and 82% accu-
may have a similar sensitivity to identify ICH, but racy to predict hematoma expansion when the score
is > 3 (Table 2, Fig. 5) [10].
Fig. 2  Typical locations for intracerebral hemorrhage (ICH). ICH due to chronic hypertension is usually due to rupture of small penetrating arterioles
and typically occurs in the basal ganglia (a), thalamus (b), cerebellum (d), and pons (e). ICH from cerebral amyloid angiopathy and sympathomi-
metic drugs of abuse such as cocaine or methamphetamine often occurs in lobar regions such as the temporal lobe (c). Supratentorial ICH would
be considered as basal ganglia, thalamic, or lobar (a–c), whereas ICH originating in the cerebellum or pons would be considered infratentorial (d–e).
a, b, and e also demonstrate IVH

Management evaluation. In general, if an ICH patient is comatose,


Initial Patient Assessment and Primary Intervention: ABCs rapid sequence intubation (RSI) should be undertaken,
and the ICH Score with a goal of normoventilation (see the ENLS Airway,
Ventilation, and Sedation protocol).
After Arrival in Hospital An initial clinical assessment of the patient’s condi-
Until the diagnosis of ICH is made from neuroimag- tion and stroke severity is essential for rapid treat-
ing, overall airway and hemodynamic management ment planning and communication among providers.
proceeds in a common pathway with other stroke sub- While performance of a complete, detailed neurological
types. However, immediately following the ICH diag- examination is ideal, much information can be gleaned
nosis, disease-specific treatment can be instituted. from a quick assessment using existing clinical grad-
Airway management, if not already dealt with appro- ing scales. The ICH score is the most commonly used
priately in the prehospital phase (specifically the need validated clinical grading scale for patients with ICH,
for intubation for airway protection), should be con- combining elements related to patient demographics,
sidered immediately after arrival. Thus, while “air- clinical condition, and neuroimaging findings that are
way” is listed under secondary treatment in the ENLS readily available at the time of hospital admission [11,
ICH algorithm (Fig. 1), it is concurrent with the initial
Fig. 3  ABC/2 method for estimating ICH hematoma volume [5].
Right basal ganglia ICH. The axial CT image with the largest cross- Fig. 4  Contrast extravasation (“spot sign”) in acute ICH. In this post-
sectional area of hemorrhage is selected and is the reference slice. contrast image obtained after administration of IV contrast during a
In this example, the largest diameter (a) is 6 cm, the largest diameter “code stroke” CT (non-contrast head CT, CT angiogram, CT perfusion),
perpendicular to (a) on the same slice (b) is 3 cm, and hemorrhage contrast extravasation is present in this acute left temporal lobe ICH.
is seen on 6 slices of 0.5 cm (5 mm) thickness for a (c) of 3 cm (not This finding is commonly referred to as a “spot sign” (arrows) and is
shown). Note that for (c), if the hematoma area on a slice is approxi- associated with increased risk of hematoma expansion
mately 25–75% of the hematoma area on the reference slice used
to determine (a), then this slice is considered half a hemorrhage
slice, and if the area is less than 25% of the reference slice, the slice is
not considered a hemorrhage slice. Thus, the hematoma volume is
Table 2  The BAT score
(6 × 3 × 3)/2 = 27 cc. Alternately, (C) can be assessed by measuring
the largest diameter, superior to inferior, that is seen on coronal or Individual components of the BAT score
sagittal images. Multiply (a) times (b) times (c), then divide by 2 to Variable Points
obtain the hematoma volume
Blend sign
 Present 1
12]. Several other useful clinical grading scales are also  Absent 0
available [13–15]. Any hypodensity
Components of the ICH score include age, initial  Present 2
Glasgow Coma Scale (GCS) score, hematoma volume,  Absent 0
hematoma location (supratentorial or infratentorial), Time from onset to NCCT​
and presence of intraventricular hemorrhage (IVH).  <2.5 h 2
Table 3 demonstrates the components of the ICH score,  ≥2.5 h or unknown 0
with the full score being the sum of points given for NCCT indicates non-contrast computed tomography
each component. Each point increase in the ICH score The blend sign is defined as a hypoattenuating area next to a hyperattenuating
is associated with an increased risk of mortality and area of the hematoma, with sharp separation between the 2 regions and a
density difference of at least 18 Hounsfield units
a decreased likelihood of good functional outcome.
Intrahematoma hypodensity is defined as a hypodense region inside the
The ICH score is best used as a communication tool hemorrhage with any shape and dimension and lack of connection with the
among providers and with patients or family members surrounding brain parenchyma
regarding a patient’s condition rather than as a tool to
precisely prognosticate outcome. While it is tempt-
ing to utilize clinical grading scales to triage severely
impaired patients toward less-aggressive intervention,
this approach is not recommended. Rather, in general,
initial aggressive therapy is recommended in order to
avoid the potential for a self-fulfilling prophecy of poor
outcome in the context of early care limitations [1, 16,
17].

Primary Intervention: Blood Pressure, Coagulopathy,


and Surgery
Following the diagnosis of ICH, immediate consideration
should be given to the need for (a) acute control of ele-
vated blood pressure, (b) correction of coagulopathy due
to medications or underlying medical conditions, and (c)
the need for urgent surgical hematoma evacuation. These
are common themes that should form part of the initial
ICH evaluation and treatment plan. Decisions regarding
these interventions will influence the succeeding aspects
Fig. 5  Non-contrast head CT demonstrating blend sign. This non-
contrast CT was performed 1.5 h after symptom onset and shows the
of ICH care, such as disposition from the ED, planning
presence of the blend sign (black arrow) which is defined as areas for repeat imaging, and need for ICP monitoring or con-
within the ICH of differing densities with a sharp separation between tinuous electroencephalography (cEEG).
the regions and intrahematoma hypodensity (red arrow). The BAT Hematoma expansion is common in patients with acute
score that is calculated from this CT is 5: 2 for time of onset, 1 for ICH, and this is associated with worsened outcomes [6,
blend sign and 2 for hypodensity (Color figure online)
18]. Though the pathophysiology that leads to hematoma
expansion is incompletely understood, it tends to occur
early (within a few hours of onset) and coagulopathy
Table 3  The ICH score [10] increases the frequency of its occurrence and its extent
Component ICH [19]. However, hematoma expansion is common even
score in patients without coagulopathy or who are not receiv-
points ing antithrombotic medications. Thus, intervention to
Glasgow Coma Scale address treatable aspects should not be delayed pending
 3–4 2 patient disposition.
 5–12 1
 13–15 0 Blood Pressure Management
ICH volume (cc) Elevated blood pressure is extremely common in patients
  ≥ 30 1 with acute ICH. While it seems intuitive that elevated
  < 30 0 blood pressure may predispose to hematoma expansion
Presence of IVH due to increased bleeding or to elevated ICP from wors-
 Yes 1 ening edema, clinical studies have had conflicting results
 No 0 regarding the impact of acutely elevated blood pressure
Infratentorial origin of ICH and the value of acutely lowering the blood pressure [20,
 Yes 1 21]. There has been a concern that acutely lowering blood
 No 0 pressure could lead to ischemic brain injury in the peri-
Age (years) hematoma region, but this risk has not been supported
  ≥80 1 by recent studies, at least for smaller ICH [22, 23].
  < 80 0 While blood pressure management has remained con-
Total ICH score 0–6 troversial, current approaches favor rapid lowering of
moderately elevated blood pressures [1, 2]. Two pilot
30-Day mortality: ICH score: 0–0%; 1–13%; 2–26%; 3–72%; 4–97%; 5–100%
randomized clinical trials, INTERACT and ATACH,
suggested that acutely lowering systolic blood pres-
sure (SBP) to below 140  mmHg is safe [24, 25]. These
were followed by pivotal phase III efficacy trials to test
the impact of blood pressure lowering on clinical out- Alternatively esmolol can be given as an infusion at
come. INTERACT-2 was a phase III clinical trial of acute 50–300 mcg/kg/min. For those patients with baseline
blood pressure lowering in ICH patients presenting with bradycardia, hydralazine as a bolus 10–20  mg every
a SBP between 150 and 200  mmHg [26]. Patients were 4–6  h may be used. Nicardipine is a calcium channel
randomized to two different SBP thresholds: a stand- blocker of the dihydropyridine family that is more selec-
ard threshold of < 180 mmHg and an intensive threshold tive for coronary and cerebral vascular beds. A common
of < 140  mmHg for the initial 7  days after ICH occur- initial nicardipine dose of 2.5–5 mg/h is often used, with
rence. Patients in the intensive arm had modestly bet- up-titration every 15 min as needed up to 15 mg/h. Cle-
ter outcomes with about 3% fewer patients having vidipine is another calcium channel blocker that acts
death or severe disability (defined as a modified Rankin even more rapidly than nicardipine. It can be initiated at
Scale score of 3–6). Interestingly, there was no differ- 1–2  mg/h, and doubling the dose every 1990  s, but not
ence in hematoma expansion between groups. ATACH to exceed 32  mg/h. Other intravenous antihyperten-
2 had a similar design and tested the two SBP thresh- sive medications including urapidil and enalaprilat may
olds of < 180 and < 140  mmHg for the initial 24  h after be used depending on local availability and practices.
ICH using the titratable intravenous agent nicardipine If possible, nitroprusside should be avoided due to its
[27]. ATACH 2 did not demonstrate a difference in out- potential for cerebral vasodilation, disturbed cerebral
come between treatment groups. However, there was an autoregulation, and elevated ICP. ICU admission is rec-
increased risk of renal adverse events in the group with ommended, due to the close monitoring and frequent
SBP < 140 mmHg. (Most patients in the aggressive blood medication changes required to lower blood pressure. A
pressure arm of this trial had a minimum SBP close to more detailed discussion of common antihypertensive
120 mmHg.) medications utilized in neurological emergencies can be
The 2015 American Heart Association/American found in the ENLS Pharmacotherapy chapter. Invasive
Stroke Association Guidelines for the Management of arterial monitoring is not mandatory but will facilitate
ICH recommend that it is safe to acutely lower SBP to titration and improve safety of antihypertensive medica-
140  mmHg in patients presenting with SBP between tion infusion.
150 and 220  mmHg. And for those presenting with
SBP > 220 mmHg, it is reasonable to consider aggressive Coagulopathy: Anticoagulants, Antiplatelet Agents,
reduction in blood pressure with continuous intrave- and Heparin
nous antihypertensive medication infusion and frequent The use of antithrombotic medications for prevention
BP monitoring (e.g., invasive BP monitoring). Thus, it and treatment of ischemic stroke, cardiovascular dis-
appears reasonable to target a SBP between 140 and ease, and systemic venous thromboembolism is common
180 mmHg with the specific threshold determined based and is increasing as the population ages. Antithrombotic
on patient comorbidities and level of chronic hyperten- medications are a risk factor for the occurrence of ICH,
sion. Although the clinical difference between these two as well as for hematoma expansion if an ICH occurs.
SBP thresholds may be modest and debatable, none of Given the range of antithrombotic medications, includ-
the current guidelines recommend allowing blood pres- ing warfarin, heparin, antiplatelet agents such as aspirin
sure to remain extremely elevated without treatment and clopidogrel, and newer agents such as dabigatran,
[1, 2]. Acute lowering of blood pressure is reasonable in rivaroxaban, and apixaban, the specific risks and inter-
patients presenting with more extreme levels of hyper- ventions to reverse coagulopathy vary. Additionally,
tension, but less is known about the specific safety and coagulopathies may be due to underlying medical condi-
efficacy of treatment [1]. tions, such as liver disease or hematologic malignances.
Basic principles of blood pressure lowering in ICH are The second focus in ICH management is on correc-
that management should be initiated immediately and a tion of coagulopathy. As part of the initial evaluation
titratable agent should be used to ensure that the target of the ICH patient, a medical history and medication
value is reached quickly and with minimal potential for list should be obtained from the patient, family, pre-
overshoot. IV beta blockers and calcium channel block- hospital providers, and/or medical record; specifically
ers are the most commonly used medications for this the use of antithrombotic medication and, if possible,
indication in the ED and the intensive care unit (ICU). when the last dose was taken should be noted. Urgent
Labetalol is rapid acting, has mixed alpha- and beta- laboratory tests should include a complete blood count
adrenergic antagonism, and is commonly used in the ED (CBC) with platelet count, an international normalized
in an initial bolus dose of 5–20 mg This can be repeated ratio (INR), and a partial thromboplastin time (PTT). A
every 5–10  min up to 300  mg. It can also be adminis- general principle is that any ICH occurring in a patient
tered as an infusion (0.5–2.0  mg/min) titrated to effect. on antithrombotic medications should be considered
life-threatening due to the risk of hematoma expansion. FFP only if PCC is not available) with the dose adjusted
Interventions to correct coagulopathy are based on this based on INR [30]. However, the specific dose may vary
history and laboratory information more than on size based on the PCC formulation used at a specific hospi-
or location of the hematoma or clinical scores. tal. Current guidelines [1, 30] recommend the use of vita-
Patients taking a vitamin K antagonist such as war- min K 10 mg administered intravenously by slow push, in
farin and whose INR is > 1.4 should receive agents to conjunction with another more rapidly acting agent (e.g.,
normalize the INR to ≤1.4. (For the purpose of emer- PCC), as it typically takes hours after vitamin K adminis-
gent EVD placement, no INR threshold has been estab- tration for reversal of warfarin-induced coagulopathy, but
lished, and an INR of ≤ 1.5 is generally considered safe it has a more long-lasting effect than PCC or FFP [29].
[28].) Options have included the administration of fresh While rFVIIa also quickly reverses an elevated INR,
frozen plasma (FFP), vitamin K, prothrombin complex this may reflect a specific effect on the INR laboratory
concentrates (PCC), and the hemostatic agent recom- test and a clinically important coagulopathy may remain.
binant factor VIIa (rFVIIa) although PCC is now the rFVIIa has been shown to decrease hematoma growth in
recommended approach [29, 30]. The most important non-coagulopathic ICH patients, but this did not trans-
principle is to normalize the INR as soon as possible, late into improved clinical outcome [35]. Thus, rFVIIa
ideally within minutes. is not recommended for use in ICH patients with or
While FFP is widely used for reversing the effect of without warfarin-related coagulopathy [1]; however, it is
warfarin, it may not be optimal in other medical con- occasionally used in patients with coagulopathy related
ditions. FFP contains factors I (fibrinogen), II, V, VII, to liver failure.
VIII, IX, X, XI, XIII, and antithrombin. Fairly large vol- Observational studies have varied regarding the impact
umes of FFP (10–15  ml/kg) are often required for full of concurrent antiplatelet therapy on hematoma expan-
reversal of anticoagulation, and this places patients at sion and outcome for patients presenting with ICH,
risk of volume overload and pulmonary edema [31]. though increased risk of hematoma growth while on
FFP, like other blood products, also carries a risk of these agents is suggested [36–39]. There has been het-
transfusion-related events and requires thawing after erogeneity in clinical practice, ranging from the empiri-
cross-matching by a blood bank. cal use of platelet transfusions, to determining the need
PCCs contain factors II, IX, X (and varying amounts for transfusion by laboratory tests for platelet func-
of VII, depending on the specific preparation) with tion, to complete avoidance of platelet transfusions. The
much higher concentrations of clotting factors in PATCH study was an open-label clinical trial testing
smaller amounts of volume than FFP. The term four- the efficacy and safety of platelet transfusion in patients
factor PCC is used to designate that sufficient concen- with ICH occurring while on an antiplatelet agent for at
trations of factors II, VII, IX, and X are present in the least a week [40]. Platelet transfusions did not improve
specific preparation. PCCs can correct the INR within outcome and were associated with a significant increase
minutes, faster than FFP, and with fewer cardiopulmo- in risk of death and more adverse events. Thus, platelet
nary complications [32]. In a prior observational study transfusion is not recommended for most patients with
comparing PCC and FFP, there was no difference in ICH occurring while on an antiplatelet agent [30]. Few
hematoma growth in patients whose INR was corrected patients in PATCH were on clopidogrel, and those under-
within 2 h [33], suggesting that the timing of coagulop- going neurosurgical procedures were excluded. The most
athy reversal, not the specific agent, makes the greatest recent antithrombotic reversal guidelines from NCS rec-
impact. However, the recent INCH clinical trial dem- ommend platelet transfusion for patients on antiplatelet
onstrated the superiority of four-factor PCC in a dose medications who are undergoing a neurosurgical pro-
of 30  IU/kg over FFP 20  ml/kg in rapidly reversing an cedure [30]. They also recommend considering a single
elevated INR to ≤ 1.2 in 54 ICH patients on a vitamin K intravenous dose of 0.4 mcg/kg of DDAVP (desmopres-
antagonist [34]. Furthermore, hematoma expansion was sin) in antiplatelet medication-related ICH. Additional
less in patients receiving PCC. There was a trend for trials are assessing platelet transfusion in ICH patients
lower mortality and better functional outcome in the as well as the role of platelet function assays in directing
PCC-treated patients; however, the study was stopped treatment.
early by its regulatory oversight body because of the Newer anticoagulants, such as direct thrombin inhibi-
finding of less hematoma expansion in the PCC group tors (e.g., dabigatran) or direct Xa inhibitors (e.g., rivar-
and therefore was underpowered to formally assess a oxaban and apixaban), now have specific reversal agents
clinical outcome difference. available. Idarucizumab is a targeted monoclonal anti-
The most recent Neurocritical Care Society (NCS) body that binds to the thrombin binding site of dabi-
guidelines recommend weight-based dosing for PCC (or gatran [41]. It is approved for use and is recommended
as the initial reversal agent for patients with ICH while Surgical Hematoma Evacuation
on dabigatran [30]. Activated charcoal (50 gm) should Though most patients with acute ICH do not require
also be given if ICH occurs within 2  h of the most surgery for removal of the hematoma, it is worthwhile
recent dabigatran dose. Less-recommended alterna- to address the option of surgery immediately after ICH
tives for reversal of direct thrombin inhibitors if idaru- diagnosis, since the theoretical benefits of surgery
cizumab is not available are the activated PCC FEIBA include prevention of brain herniation, improvement in
(factor VIII inhibitor bypassing activity) or four-factor elevated ICP, and removal of blood and blood degrada-
PCC; however, these approaches have not been formally tion products that may produce cytotoxic and vasogenic
tested and do not fully reverse dabigatran coagulopathy secondary brain injury.
[30, 42, 43]. As of 2017, FDA has approved Andexanet After decades of ambiguity, the effects of surgical
Alfa for reversal of rivaroxaban and apixaban, based on evacuation were addressed in the Surgical Trial in Intrac-
ANNEXA-4 trial, the final report of which included 352 erebral Hemorrhage (STICH) that found early surgical
patients with active bleeding (227 with ICH) [44]. The evacuation of a supratentorial ICH was not harmful, but
reduction in anti-Xa activity with the administration of there was no difference in long-term mortality or func-
Andexanet Alfa correlated with clinical hemostatic effi- tional outcome [47]. Because the subgroup of patients in
cacy in patients with ICH. If this is not available, it has STICH with lobar ICH within 1  cm of the cortical sur-
been suggested that PCCs may have some effectiveness face may have benefited from surgical evacuation, the
in reversing the effect of rivaroxaban and apixaban [45]. STICH II clinical trial was undertaken for this group of
The currently recommended approach is to use Andexa- patients [48]. However, STICH II did not demonstrate
net Alfa. Note that the half-life of the reversal agents may a significant benefit to early hematoma evacuation in
be shorter than the Xa inhibitors, and if available, anti- these patients either. The recently completed MISTIE
Xa activity should be monitored to determine whether (minimally invasive stereotactic hematoma evacuation
additional dose of Andexanet Alfa is needed. If this is not with tPA instillation) showed that the technique is safe
available, use FEIBA or four-factor PCC with the addi- and effective in reducing ICH volume faster than usual
tion of oral charcoal to retard absorption if the last dose medical management; however, despite improved mor-
of direct Xa inhibitor was within 2  h [30]. It should be tality, there was no statistically significant improvement
noted that additional laboratory tests, such as endoge- in functional outcomes, unless ICH volume was reduced
nous thrombin potential and thrombin clotting time, may to < 15 ml [49]. Minimally invasive endoscopic hematoma
have some value in assessing the activity of these newer aspiration techniques are also being studied [50–52]. At
anticoagulant agents. Vitamin K is of no value, and FFP is present, routine removal of supratentorial hematoma
of unclear utility. Currently, there is no approved reversal cannot be endorsed, but it is still undertaken as a life-
agent for edoxaban and betrixaban. Detailed information saving measure in selected patients, such as those who
on reversal of warfarin, direct thrombin inhibitors, and are deteriorating clinically and are considered salvageable
factor –Xa inhibitors can be found in the ENLS Pharma- based on characteristics such as comorbidities and age.
cotherapy module. In contrast, several case series suggest that patients
Unfractionated heparin is used for many medical con- with cerebellar ICH > 3 cm in diameter or with compres-
ditions, including acute coronary syndromes, pulmo- sion of the brain stem or hydrocephalus may benefit from
nary embolism, and endovascular surgery, as well as for surgical hematoma evacuation [53, 54]. There has not
maintaining the patency of indwelling catheters. Heparin been a randomized trial of cerebellar hematoma evacua-
binds to and activates antithrombin III, thus inactivating tion analogous to STICH, but it is not clear there is equi-
thrombin and favoring thrombolysis. The reversal agent poise to justify such a trial.
for heparin is protamine sulfate, administered 1  mg for Current American Heart Association ICH guidelines
every 100 units of heparin received in the prior 2 h, with recommend that patients with cerebellar hemorrhage
a maximum dose of 50 mg [46]. Protamine sulfate binds who are deteriorating neurologically or have brain-
to and inactivates heparin, allowing it to be broken down stem compression should undergo surgical removal
by the reticuloendothelial system. Given the short half- of the hemorrhage as soon as possible. Initial treat-
life of heparin, reversal is likely unnecessary if the last ment of these patients with ventricular drainage alone
dose was received greater than 4  h prior to ICH onset. rather than surgical evacuation is not recommended [1].
Protamine sulfate can also be used in the same dose in Supratentorial hematoma evacuation or decompressive
an attempt to reverse the effect of low molecular weight hemicraniectomy might be considered as a life-saving
heparin that was given within the prior 8 h. However, this measure in deteriorating patients. Correction of coagu-
reversal may be incomplete physiologically but may be lopathy is critical in patients undergoing surgical hema-
enough for cessation of bleeding. toma evacuation.
Secondary Intervention: Hospital Admission, ICP on current Brain Trauma Foundation Guidelines on
Management, and Seizures CPP threshold. (Note this recommendation is based on
Ideally, patients with acute ICH should be admitted to patients with severe traumatic brain injury.) Ventricu-
an ICU based on the need for close monitoring of neu- lar catheters are beneficial in their ability to both meas-
rological and hemodynamic condition and the risk of ure ICP and drain cerebrospinal fluid (CSF); therefore,
early deterioration from hematoma expansion, cerebral they should be used in patients with hydrocephalus. In
edema, hydrocephalus, or airway compromise. Admis- contrast, intraparenchymal fiberoptic monitors have a
sion to a neurological critical care unit (NCCU) has lower risk of hemorrhage and infection, but cannot be
been associated with improved outcomes compared with used to drain CSF. Correction of coagulopathy prior to
admission to a non-NCCU [55]. Acknowledging that ICP monitor insertion is desirable to minimize the risk
certain patients will require transfer between hospitals of hemorrhage. An INR ≤ 1.5 is recommended prior to
for neurological intensive care management, neurosurgi- EVD insertion.
cal intervention, or neurointerventional capabilities, all While seizures may occur in ICH patients, their inci-
aspects of ICH primary intervention can and should take dence and impact on outcome have varied across studies
place without delay in the initial presenting hospital. [63, 64]. In a single study, prophylactic anticonvulsants
Specifically, correction of coagulopathy with appropri- reduced seizure occurrence in lobar ICH [64]. However,
ate agents, blood pressure control, and treatment of acute two more recent studies found worse functional out-
seizures should be initiated in the ED of the presenting comes in patients routinely given prophylactic anticon-
hospital and not deferred until after transfer. It is critical vulsants (primarily phenytoin) [65, 66]. While comatose
that the above-discussed aspects of acute ICH evaluation ICH patients may have a high risk (approximately 20%)
and treatment are initiated at the time of original diag- of non-convulsive seizures, the impact of prophylactic
nosis and that transitions in care are smooth from ED anticonvulsants on their occurrence is also unclear [67,
to ICU (or operating room, interventional radiology, or 68]. Current guidelines do not recommend routine use
comprehensive stroke center). of prophylactic anticonvulsants [1], though some practi-
While this ENLS ICH protocol is principally concerned tioners still use a short course in patients with lobar ICH
with the initial evaluation and treatment period, it is and those undergoing surgical hematoma evacuation.
important to anticipate the health care needs of the fol- Newer antiepileptic agents such as levetiracetam are now
lowing 24–72  h as part of care planning. The first 24  h more commonly used for prophylaxis. Although antie-
is critical for blood pressure management, identification pileptic agents have been suspected to be associated with
of seizures, ICP management, and maintaining a secure worse outcome after ICH, data from the ERICH study did
airway. Avoidance of fever, hyperglycemia/hypoglyce- not support this contention as levetiracetam use was not
mia, and hypoxia are also important, as these may impact associated with poor outcome at 3  months [69]. How-
outcomes [1, 56, 57]. In addition, patients with ICH are ever, the long-term cognitive impact of AED prophylaxis
at increased risk of the development of deep venous remains unknown. Clinical seizures should be treated,
thrombosis (DVT); current guidelines recommend use and continuous EEG monitoring should be performed in
of intermittent pneumatic compression devices at hos- patients with inadequately explained decreased level of
pital admission, as well as initiation of prophylaxis-dose consciousness.
unfractionated or low molecular weight heparin within A checklist for the acute management of the ICH
1–4  days following onset (assuming cessation of bleed- patient according to the principles of ENLS is presented
ing) [1, 58]. in Table 4. This could be used as a checklist for proceed-
The incidence and impact of elevated ICP in ICH ing throughout the domains of care from prehospital, to
have received limited study, but it is undoubtedly a fac- ED, to disposition in the Neurocritical Care Unit, OR,
tor in management [59–62]. Patients with IVH are at or interfacility transfer and can be shared across medical
risk of hydrocephalus and elevated ICP. Current guide- providers as this care proceeds. Note that frequent reas-
lines for ICP monitoring in ICH follow the approach sessment of ABCs and clinical neurological status is a key
in severe traumatic brain injury, with ICP monitoring component throughout the care pathway as is revisiting
recommended in patients with GCS ≤ 8, large hema- the effectiveness of initial interventions such as blood
tomas with mass effect suggestive of elevated ICP, or pressure lowering and coagulopathy reversal in rapidly
hydrocephalus. As a goal, an ICP < 22  mmHg should achieving the desired targets.
be maintained, with a minimal cerebral perfusion pres-
sure (CPP) of 60  mmHg, adjusted based on an indi- Nursing Considerations
vidual patient’s cerebral autoregulation status [1]. A Nursing responsibilities in emergency care of ICH
CPP between 60 and 70  mmHg is reasonable, based patients are focused on monitoring and careful titration
Table 4  Standardized ICH management

Prehospital care
 □ ABCs
 □ Fingerstick glucose, peripheral IV
 □ Determine time of onset and circumstances
 □ Perform prehospital stroke screen
 □ Brief medical history and medication list
 □ Triage to stroke center
 □ Perform prehospital notification of stroke patient
ED care
 □ Emergent triage to high acuity area
 □ Perform primary assessment—ABCs
 □ Perform focused neurological examination (GCS, NIHSS)
 □ Obtain baseline screening laboratories (CBC and platelet count, electrolytes, INR and PTT, glucose)
 □ Obtain cerebrovascular imaging as soon as possible (non-con CT, stroke CT/CTA/CTP, or MRI)
 □ Obtain brief medical history and medication list
After confirmation of ICH
 □ Reassess ABCs (consider intubation if comatose, GCS < 9)
 □ Initiate blood pressure intervention (target SBP 140–180 mmHg)
 □ Quantify ICH volume (ABC/2 calculation)
 □ Perform ICH score (0–6)
 □ Begin correction of anticoagulation as required (goal INR ≤ 1.4)
 □ Consult neurosurgery for potential hematoma evacuation or ICP monitor placement
 □ Admit to (neuro) ICU (may require transfer)
In-hospital setting
 □ Continue to reassess ABCs
 □ Continue neurological reassessment
 □ ICP monitor and/or ventriculostomy for treatment of elevated ICP or hydrocephalus
 □ Continue management of blood pressure
 □ Place arterial blood pressure catheter as needed
 □ Place central venous catheter as needed
 □ Urine toxicology screen (if not already done)
 □ Foley catheter (needed for most ICH patients early)
 □ Feeding tube (goal to begin feeding within first day)
 □ DVT prophylaxis with sequential compression devices (consider heparin/LWMH after ICH stability between days 1–4)
 □ Recheck INR and PTT if patient was coagulopathic and received reversal agents
 □ No anticonvulsant prophylaxis; treat clinical seizures; continuous EEG if level of consciousness impaired out of proportion to ICH or IVH
 □ Consider need for repeat head CT
 □ Consider need for catheter cerebral angiography for etiology of ICH

of BP to meet goals. The primary nurse recognizes and Pediatric Considerations


alerts appropriate members of the emergency team with The etiology of pediatric ICH is vastly different than that
changes in cardiac rhythm, oxygenation, mental and neu- in adults because chronic hypertension and therapeutic
rological status (e.g., pupillary changes), hemodynamics, anticoagulation are less prevalent. ICH is seen much less
and ICP/CPP in a timely fashion. Lastly, activation and frequently in pediatric patients. However, children do
coordination of acute care pathways to ensure safe trans- present with life-threatening ICH due to vascular malfor-
port of patients from one level of care to the other are mations, bleeding disorders, vasculitis, sickle cell disease,
vital and best managed by the primary nurse. or infection. While less than 2% of cerebral aneurysms are
found in pediatric patients, as many as 24% of children
with intracranial aneurysms may have ICH at the time
of their initial presentation, and vascular malformations
account for almost 50% of pediatric spontaneous ICH of significant pediatric ICH are oncologic and include
[70]. Hemorrhagic transformation after ischemic stroke bleeding of brain tumors (the most common solid tumor
can occur, particularly because recognition of stroke of childhood), and ICH as a result of combined thrombo-
in children lags behind that in adults. ICH as a result of cytopenia and vascular sludging from hyperleukocytosis
hemorrhagic disease of the newborn was virtually eradi- in acute leukemic blast crisis.
cated by routine vitamin K administration after birth but A priori knowledge of your facility’s capabilities and
can be seen in cases of parental refusal. Trauma-related identification of and early referral to a center with
ICH can require emergent intervention, and while iso- expertise and comfort with neurocritically ill children is
lated ICH is uncommon compared to SDH in young chil- paramount, as optimal outcomes may be related to inter-
dren who have been abused, these children often present ventions in the first hours, and children are at risk of
without disclosure of trauma. Other important etiologies etiologies that require a variety of pediatric subspecialty

Table 5  ICH communication/hand-off


Communication—prehospital to ED (before diagnosis of ICH)

 □ Airway status—patent airway/supraglottic device/endotracheal tube


 □ Breathing—respiratory status
 □ Blood pressure and pulse
 □ Age/gender
 □ GCS, pupils
 □ Signs and symptoms
 □ Last known normal
 □ Brief relevant medical history—previous stroke, blood thinners, bleeding
 □ Current medications
Sample EMS call
This is Medic 1 with a stroke alert—85-year-old male, acute right hemiparesis, on aspirin 81 mg. Last known normal at 1300. Vital signs stable. ETA
30 min.
Communication—hand-off after ICH diagnosis has been made

 □ Age
 □ GCS, pupil exam
 □ Hematoma volume and location
 □ Other CT findings (intraventricular hemorrhage, hydrocephalus, spot sign)
 □ ICH score
 □ Airway status
 □ Blood pressure, target, and treatment initiated
 □ Coagulation parameters (INR, PT, PTT, platelet count, WBC, Hgb) and reversal treatment if any
 □ Medications given
 □ Plan for surgery if any
Sample Sign-Off Narrative
“I am signing out a 62-year-old man with known hypertension and atrial fibrillation who is presumed to be on warfarin.”
“He was found at home this morning at 9 AM by his wife who last saw him normal at 7 AM. He was talking to EMS and had left-sided weakness, GCS in
the field was 13, and BP was 170/100.”
“On arrival to the ED here, he was the same, so we drew labs and sent him for a head CT.”
“CT completed at 10 AM showed a 20 ml right thalamic ICH with mild IVH, but no hydrocephalus. There is about 4 mm of right-to-left midline shift.
CTA/CTP showed no AVM or aneurysm, but there is a positive spot sign.”
“When he returned to the ED, he was sleepier, with a GCS of 9, and his left-sided weakness was worse. So he has an ICH score of 2. His labs came back
with an INR of 1.9.”
“We intubated him using rocuronium and etomidate. PCC infusion of 2250 IU (estimated weight 90 kg; dose of 25 IU/kg) is going in now. He also had
10 mg of IV vitamin K.”
“Neurosurgery has been called, and they are on their way to see him. He is in ED Resuscitation Room 1, intubated and sedated now on propofol at 60
mcg/kg/min and fentanyl 50 mcg/hr. His BP is 140/85 with no other treatment.”
“They are ready to take him in Bed 2 in the NCCU in 5 min. Nursing is also calling report.”
care. As an example, a child with leukemic blast crisis Transport Considerations
may require emergent white cell reduction with pher- This section addresses patient transport, e.g., from ED
esis and chemotherapy in addition to management to radiology suite, ED to ICU, or interhospital trans-
of ICH and this will usually require transfer to a larger port. Before the patient is transported, the responsible
pediatric referral centers. For children with significant physician should assess the condition of the patient
ICH, the same emergent care principles described ear- to determine the necessary personnel and equipment
lier in this chapter apply regarding need for establish- needed. Patients with borderline airways should be
ment of the airway and providing adequate oxygenation intubated before transport.
and maintaining blood pressure (see pediatric section in
ENLS Airway, Ventilation and Sedation). Hypotension is 1. Personnel: In addition to a transporter, this should
defined as SBP below the 5th percentile for age (SBP 5th include an experienced nurse/physician/critical care
percentile = 70 mmHg + age in years X 2). Careful atten- transport paramedic who can manage the patient’s
tion should be given to the identification of ICH in need ABCs.
of emergent surgical evacuation, and also to detection 2. Equipment: A transport monitor that can display
and treatment of seizures, which may occur in as many as blood pressure (noninvasive or invasive), ECG, pulse
21% of children with intracranial aneurysms [71]. oximetry with supplemental oxygen and/or ventilator
There are no established parameters for treatment of as indicated. If an EVD is in place, it should be clearly
hypertension in children with ICH, but an SBP target of stated whether the drain should be clamped or open,
140–180 mmHg is reasonable in older children. Future and if open, what level it should be set at.
studies may yield parameters such as a certain  % above 3. Drugs: Basic medications for blood pressure control/
5th percentile for age, but those types of studies do not support, sedation, analgesia, and osmotic agents (e.g.,
yet exist for children on a large enough scale to make mannitol or 23.4% saline as needed).
firm recommendations beyond vigorously avoiding
hypotension and ensuring adequate cerebral perfusion Communication
pressure. Blood pressure goals for hypertensive younger When communicating to an accepting or referring physi-
children with ICH are best set in consultation with an cian about a patient with ICH, consider including the key
experienced subspecialist from pediatric emergency elements listed in Table 5.
medicine, neurosurgery, critical care, or neurology.
Nicardipine is well tolerated and the recommended Clinical pearls
dose is 0.5 mcg/kg/min, titrated by 0.5 mcg/kg/min  • Hematoma expansion occurs within the first 6–12 h in up to 40%
every 15 min to a maximum of 5 mcg/kg/min. In older of patients. This can lead to neurological deterioration and airway
compromise and should be anticipated.
children (adult weight), similar dosing and titration as
 • The “spot sign” on contrast CT/CTA and the “BAT” score on non-contrast
that for adults is followed. Hydralazine is also an option CT can help to identify patients at high risk of hematoma expansion.
with a recommended starting IV dose of 0.1–0.2  mg/  • The ABC/2 method can be quickly used to measure hematoma vol-
kg/dose every 4–6 h with a maximum dose of 2 mg for ume on a CT scan.
infants and 10–20  mg for children and adolescents.  • The ICH score is best used as a communication tool between provid-
Esmolol and labetolol are reasonable alternatives and ers rather than to predict neurological outcome.
generally well tolerated. It should be noted that beta  • Admission to a neurological critical care unit with physician and nurs-
ing expertise in ICH is associated with improved ICH outcomes.
blockers in young children should be used with caution,
particularly during the first hours of resuscitation when  • Current recommendations are to lower BP to SBP between 140 and
180 mm Hg with the specific target determined by patient related
they are best avoided, as cardiac output in this age factors.
group is heart rate dependent and artificially low heart  • Urgent coagulopathy reversal should be done to minimize ICH expan-
rate can complicate identification of increased ICP in sion. Specific reversal agents are available for all oral anticoagulants.
addition to lowering blood pressure. Developmentally,  • The role of open craniotomy is limited to cerebellar hematoma with
infants have higher baseline vagal tone and are prone brainstem compression and as a life-saving measure in selected
patients with supratentorial ICH.
to bradycardia from a variety of stimuli. Infants also
 • Current recommendations do not endorse routine prophylactic use of
have a high glucose utilization rate and are more at antiepileptics for all patients with ICH.
risk of hypoglycemia that can occur with beta blockers.  • A patient who has a level of consciousness out of proportion to imag-
Finally, while anticoagulation therapy is less common in ing findings should be investigated for subclinical seizures.
children, pediatric patients with ICH may present with
coagulation abnormalities that require careful evalu-
ation and treatment to prevent hematoma expansion
and facilitate surgical therapy.
Author details 14. Rost NS, Smith EE, Chang Y, et al. Prediction of functional outcome in
1
 Department of Anesthesiology, University of California, 200W Arbor St, San patients with primary intracerebral hemorrhage: the FUNC score. Stroke.
Diego, CA 94110, USA. 2 Department of Neurology, University of California, San 2008;39(8):2304–9.
Francisco, CA, USA. 3 Division of Critical Care Medicine, Department of Pediat- 15. Tuhrim S, Horowitz DR, Sacher M, Godbold JH. Validation and comparison
rics, Virginia Commonwealth University, Richmond, VA, USA. of models predicting survival following intracerebral hemorrhage. Crit
Care Med. 1995;23(5):950–4.
Acknowledgements 16. Hemphill JC 3rd, Newman J, Zhao S, Johnston SC. Hospital usage of early
The authors are grateful for the contributions and insight provided by the fol- do-not-resuscitate orders and outcome after intracerebral hemorrhage.
lowing reviewers: Christina Watford, BSN, RN, CCRN; Megan Corry, EdD, EMTP; Stroke. 2004;35(5):1130–4.
Jeffrey Fong, PharmD, BCPS; and Gentle Shrestha, MD. 17. Hemphill JC 3rd, White DB. Clinical nihilism in neuroemergencies. Emerg
Med Clin North Am. 2009;27(1):27–37, vii-viii.
18. Davis SM, Broderick J, Hennerici M, et al. Hematoma growth is a deter-
Publisher’s Note minant of mortality and poor outcome after intracerebral hemorrhage.
Springer Nature remains neutral with regard to jurisdictional claims in pub- Neurology. 2006;66(8):1175–81.
lished maps and institutional affiliations. 19. Flibotte JJ, Hagan N, O’Donnell J, Greenberg SM, Rosand J. Warfarin,
hematoma expansion, and outcome of intracerebral hemorrhage. Neu-
rology. 2004;63(6):1059–64.
20. Jauch EC, Lindsell CJ, Adeoye O, et al. Lack of evidence for an association
between hemodynamic variables and hematoma growth in spontane-
ous intracerebral hemorrhage. Stroke. 2006;37(8):2061–5.
21. Kazui S, Minematsu K, Yamamoto H, Sawada T, Yamaguchi T. Predisposing
References factors to enlargement of spontaneous intracerebral hematoma. Stroke.
1997;28(12):2370–5.
22. Qureshi AI, Wilson DA, Hanley DF, Traystman RJ. No evidence for an
* Important papers
ischemic penumbra in massive experimental intracerebral hemorrhage.
** Landmark papers
Neurology. 1999;52(2):266–72.
23. Zazulia AR, Diringer MN, Videen TO, et al. Hypoperfusion without
1. **Hemphill JC 3rd, Greenberg SM, Anderson CS, et al. Guidelines for the
ischemia surrounding acute intracerebral hemorrhage. J Cereb Blood
management of spontaneous intracerebral hemorrhage: a guideline
Flow Metab. 2001;21(7):804–10.
for healthcare professionals From the American Heart Association/
24. Antihypertensive treatment of acute cerebral hemorrhage. Crit Care Med.
American Stroke Association. Stroke. 2015;46(7):2032–60. Latest guideline
2010;38(2):637–48.
on the management of all aspects of spontaneous intracerebral hemor-
25. Anderson CS, Huang Y, Wang JG, et al. Intensive blood pressure reduction
rhage with comprehensive review of the literature and evidence for the
in acute cerebral haemorrhage trial (INTERACT): a randomised pilot trial.
recommendations.
Lancet Neurol. 2008;7(5):391–9.
2. Steiner T, Al-Shahi Salman R, Beer R, et al. European Stroke Organisation
26. **Anderson CS, Heeley E, Huang Y, et al. Rapid blood-pressure lower-
(ESO) guidelines for the management of spontaneous intracerebral
ing in patients with acute intracerebral hemorrhage. N Engl J Med.
hemorrhage. Int J Stroke. 2014;9(7):840–55.
2013;368(25):2355–65. INTERACT 2-Multi-center trial that demonstrates the
3. Chalela JA, Kidwell CS, Nentwich LM, et al. Magnetic resonance imag-
feasibility and safety of acute lowering of blood pressure in ICH.
ing and computed tomography in emergency assessment of patients
27. *Qureshi AI, Palesch YY, Barsan WG, et al. Intensive blood-pressure
with suspected acute stroke: a prospective comparison. Lancet.
lowering in patients with acute cerebral hemorrhage. N Engl J Med.
2007;369(9558):293–8.
2016;375(11):1033–43. ATACH 2- Another multi-center trial that demon-
4. Fiebach JB, Schellinger PD, Gass A, et al. Stroke magnetic resonance
strates the feasibility and safety of acute lowering of blood pressure in ICH.
imaging is accurate in hyperacute intracerebral hemorrhage: a multi-
28. Bauer DF, McGwin G Jr, Melton SM, George RL, Markert JM. The relation-
center study on the validity of stroke imaging. Stroke. 2004;35(2):502–6.
ship between INR and development of hemorrhage with placement of
5. Kothari R, Brott T, Broderick J, et al. The ABCs of measuring intracerebral
ventriculostomy. J Trauma. 2011;70(5):1112–7.
hemorrhage volume. Stroke. 1996;27:1304–5.
29. Guidelines on oral anticoagulation: third edition. Br J Haematol
6. Brott T, Broderick J, Kothari R, et al. Early hemorrhage growth in patients
1998;101(2):374–87
with intracerebral hemorrhage. Stroke. 1997;28:1–5.
30. Frontera JA, Lewin JJ 3rd, Rabinstein AA, et al. Guideline for reversal of
7. Goldstein JN, Fazen LE, Snider R, et al. Contrast extravasation on CT
antithrombotics in intracranial hemorrhage: a statement for healthcare
angiography predicts hematoma expansion in intracerebral hemorrhage.
professionals from the neurocritical care society and society of critical
Neurology. 2007;68(12):889–94.
care medicine. Neurocrit Care. 2016;24(1):6–46.
8. Kim J, Smith A, Hemphill JC 3rd, et al. Contrast extravasation on CT
31. Hanley JP. Warfarin reversal. J Clin Pathol. 2004;57(11):1132–9.
predicts mortality in primary intracerebral hemorrhage. AJNR Am J
32. Sarode R, Milling TJ Jr, Refaai MA, et al. Efficacy and safety of a 4-factor
Neuroradiol. 2008;29(3):520–5.
prothrombin complex concentrate in patients on vitamin K antagonists
9. Wada R, Aviv RI, Fox AJ, et al. CT angiography “spot sign” predicts
presenting with major bleeding: a randomized, plasma-controlled, phase
hematoma expansion in acute intracerebral hemorrhage. Stroke.
IIIb study. Circulation. 2013;128(11):1234–43.
2007;38(4):1257–62.
33. Huttner HB, Schellinger PD, Hartmann M, et al. Hematoma growth and
10. *Morotti A, Dowlatshahi D, Boulouis G, et al. Predicting intracerebral
outcome in treated neurocritical care patients with intracerebral hemor-
hemorrhage expansion with noncontrast computed tomography: the
rhage related to oral anticoagulant therapy: comparison of acute treat-
BAT score. Stroke. 2018;49(5):1163–9. Potential use of the routine CT scan
ment strategies using vitamin K, fresh frozen plasma, and prothrombin
and BAT score to predict hematoma expansion.
complex concentrates. Stroke. 2006;37(6):1465–70.
11. **Hemphill JC 3rd, Bonovich DC, Besmertis L, Manley GT, Johnston SC.
34. Steiner T, Poli S, Griebe M, et al. Fresh frozen plasma versus prothrombin
The ICH score: a simple, reliable grading scale for intracerebral hemor-
complex concentrate in patients with intracranial haemorrhage related
rhage. Stroke. 2001;32(4):891–7. The most frequently used scale to grade
to vitamin K antagonists (INCH): a randomised trial. Lancet Neurol.
and standardize the severity of ICH.
2016;15(6):566–73.
12. Hemphill JC 3rd, Farrant M, Neill TA Jr. Prospective validation of the ICH
35. Mayer SA, Brun NC, Begtrup K, et al. Efficacy and safety of recombinant
Score for 12-month functional outcome. Neurology. 2009;73(14):1088–94.
activated factor VII for acute intracerebral hemorrhage. N Engl J Med.
13. Broderick JP, Brott TG, Duldner JE, Tomsick T, Huster G. Volume of intrac-
2008;358(20):2127–37.
erebral hemorrhage. A powerful and easy-to-use predictor of 30-day
36. Foerch C, Sitzer M, Steinmetz H, Neumann-Haefelin T. Pretreatment with
mortality. Stroke. 1993;24(7):987–93.
antiplatelet agents is not independently associated with unfavorable
outcome in intracerebral hemorrhage. Stroke. 2006;37(8):2165–7.
37. Naidech AM, Bernstein RA, Levasseur K, et al. Platelet activity and out- 53. Firsching R, Huber M, Frowein RA. Cerebellar haemorrhage: management
come after intracerebral hemorrhage. Ann Neurol. 2009;65(3):352–6. and prognosis. Neurosurg Rev. 1991;14(3):191–4.
38. Sansing LH, Messe SR, Cucchiara BL, et al. Prior antiplatelet use does 54. Kirollos RW, Tyagi AK, Ross SA, van Hille PT, Marks PV. Management of
not affect hemorrhage growth or outcome after ICH. Neurology. spontaneous cerebellar hematomas: a prospective treatment protocol.
2009;72(16):1397–402. Neurosurgery. 2001;49(6):1378–86 discussion 86-7.
39. Thompson BB, Bejot Y, Caso V, et al. Prior antiplatelet therapy and out- 55. Diringer MN, Edwards DF. Admission to a neurologic/neurosurgical inten-
come following intracerebral hemorrhage: a systematic review. Neurol- sive care unit is associated with reduced mortality rate after intracerebral
ogy. 2010;75(15):1333–42. hemorrhage. Crit Care Med. 2001;29(3):635–40.
40. Baharoglu MI, Cordonnier C, Al-Shahi Salman R, et al. Platelet transfusion 56. Schwarz S, Hafner K, Aschoff A, Schwab S. Incidence and prognostic
versus standard care after acute stroke due to spontaneous cerebral significance of fever following intracerebral hemorrhage. Neurology.
haemorrhage associated with antiplatelet therapy (PATCH): a randomised, 2000;54(2):354–61.
open-label, phase 3 trial. Lancet. 2016;387(10038):2605–13. 57. Vespa PM. Intensive glycemic control in traumatic brain injury: what is
41. *Pollack CV Jr, Reilly PA, Eikelboom J, et al. Idarucizumab for dabigatran the ideal glucose range? Crit Care. 2008;12(5):175.
reversal. N Engl J Med. 2015;373(6):511–20. This study establishes the 58. Nyquist P, Bautista C, Jichici D, et al. Prophylaxis of venous thrombosis in
efficacy of reversal of effects of dabigatran with idarucizumab. neurocritical care patients: an evidence-based guideline: a statement for
42. Dager WE, Gosselin RC, Roberts AJ. Reversing dabigatran in life-threaten- healthcare professionals from the Neurocritical Care Society. Neurocrit
ing bleeding occurring during cardiac ablation with factor eight inhibitor Care. 2016;24(1):47–60.
bypassing activity. Crit Care Med. 2013;41(5):e42–6. 59. Chambers IR, Banister K, Mendelow AD. Intracranial pressure within a
43. Lazo-Langner A, Lang ES, Douketis J. Clinical review: clinical management developing intracerebral haemorrhage. Br J Neurosurg. 2001;15(2):140–1.
of new oral anticoagulants: a structured review with emphasis on the 60. Fernandes HM, Siddique S, Banister K, et al. Continuous monitoring of
reversal of bleeding complications. Crit Care. 2013;17(3):230. ICP and CPP following ICH and its relationship to clinical, radiological and
44. *Connolly SJ, Crowther M, Eikelboom JW, et al. Full study report of andex- surgical parameters. Acta Neurochir Suppl. 2000;76:463–6.
anet alfa for bleeding associated with factor Xa inhibitors. N Engl J Med. 61. Kamel H, Hemphill JC 3rd. Characteristics and sequelae of intracra-
2019;380(14):1326–35. This study establishes the clinical efficacy of Andexa- nial hypertension after intracerebral hemorrhage. Neurocrit Care.
net for the reversal of rivaroxaban or apixaban induced coagulopathy. 2012;17(2):172–6.
45. Eerenberg ES, Kamphuisen PW, Sijpkens MK, et al. Reversal of rivaroxaban 62. Ziai WC, Torbey MT, Naff NJ, et al. Frequency of sustained intracranial
and dabigatran by prothrombin complex concentrate: a randomized, pressure elevation during treatment of severe intraventricular hemor-
placebo-controlled, crossover study in healthy subjects. Circulation. rhage. Cerebrovasc Dis. 2009;27(4):403–10.
2011;124(14):1573–9. 63. De Herdt V, Dumont F, Henon H, et al. Early seizures in intracerebral
46. Schulman S, Bijsterveld NR. Anticoagulants and their reversal. Transfus hemorrhage: incidence, associated factors, and outcome. Neurology.
Med Rev. 2007;21(1):37–48. 2011;77(20):1794–800.
47. Mendelow AD, Gregson BA, Fernandes HM, et al. Early surgery versus 64. Passero S, Rocchi R, Rossi S, Ulivelli M, Vatti G. Seizures after spontaneous
initial conservative treatment in patients with spontaneous supraten- supratentorial intracerebral hemorrhage. Epilepsia. 2002;43(10):1175–80.
torial intracerebral haematomas in the International Surgical Trial 65. Messe SR, Sansing LH, Cucchiara BL, et al. Prophylactic antiepileptic
in Intracerebral Haemorrhage (STICH): a randomised trial. Lancet. drug use is associated with poor outcome following ICH. Neurocrit Care.
2005;365(9457):387–97. 2009;11(1):38–44.
48. Mendelow AD, Gregson BA, Rowan EN, et al. Early surgery versus initial 66. Naidech AM, Garg RK, Liebling S, et al. Anticonvulsant use and outcomes
conservative treatment in patients with spontaneous supratentorial after intracerebral hemorrhage. Stroke. 2009;40(12):3810–5.
lobar intracerebral haematomas (STICH II): a randomised trial. Lancet. 67. Claassen J, Jette N, Chum F, et al. Electrographic seizures and periodic dis-
2013;382(9890):397–408. charges after intracerebral hemorrhage. Neurology. 2007;69(13):1356–65.
49. Hanley DF, Thompson RE, Rosenblum M, et al. Efficacy and safety of mini- 68. Vespa PM, O’Phelan K, Shah M, et al. Acute seizures after intracerebral
mally invasive surgery with thrombolysis in intracerebral haemorrhage hemorrhage: a factor in progressive midline shift and outcome. Neurol-
evacuation (MISTIE III): a randomised, controlled, open-label, blinded ogy. 2003;60(9):1441–6.
endpoint phase 3 trial. Lancet. 2019;393(10175):1021–32. 69. Sheth KN, Martini SR, Moomaw CJ, et al. Prophylactic antiepileptic drug
50. Auer LM, Deinsberger W, Niederkorn K, et al. Endoscopic surgery versus use and outcome in the ethnic/racial variations of intracerebral hemor-
medical treatment for spontaneous intracerebral hematoma: a rand- rhage study. Stroke. 2015;46(12):3532–5.
omized study. J Neurosurg. 1989;70(4):530–5. 70. Gross BA, Smith ER, Scott RM, Orbach DB. Intracranial aneurysms in the
51. Niizuma H, Shimizu Y, Yonemitsu T, Nakasato N, Suzuki J. Results of stereo- youngest patients: characteristics and treatment challenges. Pediatr
tactic aspiration in 175 cases of putaminal hemorrhage. Neurosurgery. Neurosurg. 2015;50(1):18–25.
1989;24(6):814–9. 71. Garg K, Singh PK, Sharma BS, et al. Pediatric intracranial aneurysms–our
52. Vespa P, McArthur D, Miller C, et al. Frameless stereotactic aspiration and experience and review of literature. Childs Nerv Syst. 2014;30(5):873–83.
thrombolysis of deep intracerebral hemorrhage is associated with reduc-
tion of hemorrhage volume and neurological improvement. Neurocrit
Care. 2005;2(3):274–81.

You might also like