The Acute Neurologi
The Acute Neurologi
Acute
Neurology
Survival Guide
A Practical Resource
for Inpatient and ICU
Neurology
Catherine S.W. Albin
Sahar F. Zafar
Editors
123
The Acute Neurology Survival Guide
Catherine S. W. Albin • Sahar F. Zafar
Editors
https://doi.org/10.1007/978-3-030-75732-8
This Springer imprint is published by the registered company Springer Nature Switzerland AG
The registered company address is: Gewerbestrasse 11, 6330 Cham, Switzerland
Foreword
v
Preface
The history of The Acute Neurology Survival Guide goes back 5 years, to
2016, when we recognized our mutual interest in improving the residents’
educational experience in the neuro-ICU.
At that time, I (Sahar) had just joined my first faculty position in the neuro-
ICU and saw that the many rotating residents and interns needed a struc-
tured curriculum and orientation. Witnessing this need, I collaborated with
the medical and nursing ICU directors and residency program director to
develop a systematized orientation and ICU curriculum.
Meanwhile, I (Casey) had just completed my first year of neurology resi-
dency. Like residents all over the country, I had spent most of the year
learning “by doing” and bouncing between various blogs, online orientations,
textbooks left in resident workrooms, and various Epic “dot phrases” to
ensure that I had a reasonable assessment and plan to present, but I never
found a centralized resource to address, in a practical way, the many
questions I had during my nights on call.
Recognizing the absence of a responding-clinician-level source for guid-
ance, we set out to create a centralized, check-list driven “how-to” guide
which we termed the “NeuroICU Survival Guide.” The first version was
printed by our neurology residency program in 2017 and in the years that
followed became the core manual for trainees and advance practice provid-
ers working in the neuro-ICU.
Since that time, with the help and advice of pharmacists, residents, fellows,
and APPs, we have dramatically expanded the content – adding chapters on
common inpatient issues and routine consult questions – but the goal has
always been the same: to create an incredibly easy-to-use, visually acces-
sible how-to manual that covers exactly what every clinician needs to know
to care for the patient in front of them.
vii
viii Preface
Inside you will find checklists, scoring systems, pro-tips, helpful reminders,
as well as concise summary of the pertinent literature. We have included
over 150 images, charts, and diagrams in an effort to distill complex topics
into understandable learning points that are accessible even at 4 AM or at
the end of a 30-hour call.
In Part I, you will find tangible guidance about how to pre-round, structure a
presentation, examine neurologically ill patients, and interpret the core
diagnostics obtained in many neurology patients: CT, MRI, and EEG.
Parts II and III contain chief-complaint oriented chapters that delve into the
care of vascular and non-vascular inpatient admissions. Part IV covers the
topics central to caring for Neuro-Medicine and Neurosurgery patients in the
neuro-ICU.
Part V is a compilation of commonly referenced resources in a useable
format: sections of the brainstem that are oriented the way you would see
them in a radiology image, common drug-drug interactions to be aware of,
and a comprehensive guide to anti-epileptic drugs (AEDs).
As neurology and neurosurgery are rapidly evolving fields, each year there
are hundreds of new studies that refine and transform the care of these
complex patients. We have made every effort to include the latest guide-
lines, terminology, and publications.
This work would not have been possible without the dedication and collec-
tive expertise of the residents, fellows, pharmacists, and APPs that contrib-
uted and the guidance of experienced clinicians who made this work
possible. We are deeply grateful for the time and energy that each author
contributed and acknowledge that their input has made this book stronger.
We would especially like to thank Dr. Aaron Berkowitz and Megan Barra,
PharmD for their insightful revisions and thoughful feedback; the value of
their input cannot be overstated.
Whether you are a fledging clinician or an experienced provider, we hope
that The Acute Neurology Survival Guide will improve your care of each
patient encountered in the ICU, wards, ED, or other acute care settings.
Sincerely,
ix
x Contents
Part IV NeuroICU
33 Intracranial Pressure: Theory and Management Strategies ����������������� 187
Melissa Bentley and Catherine S. W. Albin
34 Management of External Ventricular Catheters������������������������������������� 197
Catherine S. W. Albin and Sahar F. Zafar
35 Malignant Middle Cerebral Artery Infarction����������������������������������������� 199
Catherine S. W. Albin and Sahar F. Zafar
36 Intraparenchymal Hemorrhage����������������������������������������������������������������� 205
Catherine S. W. Albin and Sahar F. Zafar
37 Intracranial Hemorrhage – Landmark Trials����������������������������������������� 211
Catherine S. W. Albin and Sahar F. Zafar
38 Reversal of Selected Antithrombotics������������������������������������������������������� 215
Catherine S. W. Albin and Megan E. Barra
39 An In-Depth Review of Reversing Direct Factor
XA-Inhibitor-Related Hemorrhages��������������������������������������������������������� 221
Megan E. Barra
xii Contents
xv
xvi Contributors
First and foremost, know how you will communicate with the team. Many hospitals
use pagers, encrypted texting services, special mobile phones, team huddle, etc. It is
critically important that for any rotation you determine how you will reach the bedside
nurse, PT, OT, SLP, nutrition, and case management, and how they will reach you.
You may also need special passwords to access EEG, radiology, telemetry, etc.
Ask early.
4
PRE-ROUNDING AND PRESENTING NEUROICU
PATIENTS
Catherine S. W. Albin and Sahar F. Zafar
As for floor patients, it is critically important that for any rotation you determine how
you will reach the bedside nurse, PT, OT, SLP, nutrition and case management, and
how they will reach you.
You may also need special passwords to access EEG, radiology, telemetry, etc. Ask early!
Sedation:
Propofol 5–80 mcg/kg/min
Dexmedetomidine 0.2–1.5 mcg/kg/hour
Fentanyl 25–250 mcg/hour OR 0.5–3 mcg/kg/hour
Midazolam 1–5 mg/hour
Anti-Hypertension:
Clevidipine 1–16 mg/hour
Labetalol 1–4 mg/hour
Nicardipine 2.5–15 mg/hour
Pressors:
Norepinephrine (commonly called “levo”):
• 0–50 mcg/min, IV, continuous
• 0.01–1 mcg/kg/min, if weight based
Phenylephrine (commonly called “neo”):
• 0–300 mcg/min, IV, continuous
• Or 0.1–3 mcg/kg/min, if weight based
6
CV
• SBP goal, patient’s range
• Pressors or Ant-HTN infusions
• If patient is in shock, address in this section
• EKG, ECHO, and telemetry data, as applicable
PULM
• Ventilator settings and recent ABG and EtCO2
• CXR results
• Diuresis plan, if needed for pulmonary edema
GI
• If the patient is on TFs and if they are at goal
• Last bowel movement
RENAL
• Ins/outs
• Sodium goal and how it is being addressed
• Any CKD or AKI, and how it is being managed: HD, CRRT, monitoring K, renal
diet, etc.
ID
• Tmax, white blood cell count
• Antibiotics as ordered and how many days they have had/are planned
HEME
• Hgb level
• Plts, if significantly high or low
• Coags if they are important to the patient’s neurologic issue
ENDOCRINE
• Blood sugars if DM, baseline HgbA1c
• Treatment for hypothyroidism, if needed
• Diabetes insipidus management (may be covered in renal with sodium
management)
• Adrenal insufficiency treatment, as applicable
MSK/ONC/SKIN: as important or needed
PROPHYLAXIS/ICU Checklist
• Review of peripheral and central access
• DVT prophylaxis plan (with anti-Xa levels or PTT, as needed/monitored)
• GI prophylaxis (often with H2 blockers or PPIs), not needed for all patients, but
should be considered for ventilated patients, patients with coagulopathy, or
patients that are expected to have an extended NPO period.
• Foley (with the goal of always removing, unless it is needed for monitoring of
critical I/Os)
7
THE COMA EXAM
Catherine S. W. Albin and Sahar F. Zafar
In general, all ICU exams should include mental status, cranial nerves, motor
responses, and reflexes. Patients that were admitted with stroke should be tracked
using the NIHSS, found on page 351.
The Glasgow Coma Scale (GCS) is a simple, effective way to communicate and
track progress.
pain or at least cross midline, if a stimulus is applied to the contralateral side (see figure 3.1)
An easy way to remember that decorticate is upward flexion of the arm is that the patient is pulling
b
Decerebrate Posturing
MENTAL STATUS
It is always best to describe what the patient can/cannot do. Below are some terms
encountered in neurologic and critically ill patient.
Encephalopathic: Confused, inattentive
Stupor: Unresponsiveness that requires vigorous/continuous stimulation.
Comatose: Cannot be aroused with vigorous stimulus
10
HERNIATION SYNDROMES
NAME PATHOLOGY SIGNS
A. Uncal Medialization of the uncas, the medial/inner most Ipsilateral fixed and dilated
Herniation part of the temporal lobe, towards the tentorium pupil
resulting in compression of the midbrain and 3rd May also have an inability to
nerve. adduct affected eye with
vestibular ocular reflex
B. Central Downward displacement of the diencephalon and Coma
Transtentorial brainstem, resulting in compression of reticular Diabetes insipidus
Herniation activating system and hypothalamus Parinaud’s syndrome (loss of
upgaze + convergence/
retraction nystagmus)
C. Falcine Displacement of the cingulate gyrus, pericallosal Contralateral leg weakness
Herniation arteries, and the ipsilateral anterior cerebral artery.
D. Tonsillar Pressure gradient across the foramen magnum Obtundation
Herniation impacting the cerebellar tonsils resulting in Hypertension
compression of 4th ventricle and brainstem, as well
as hydrocephalus.
E. Kernohan’s Compression of the contralateral cerebral peduncle Weakness that is ipsilateral to
Notch and midbrain against the tentorium cerebelli the injury
Phenomenon Contralateral pupillary
dilatation.
E
D
Figs. 3.2 and 3.3 Above is an MRI obtained on a patient who had herniated from massive ICH and
was taken to the OR for decompressive hemicraniectomy. After decompression there is evidence of
subtle DWI restriction in the lateral midbrain at the level of the cerebral peduncle (dark arrow) which
had been compressed against the cerebellar tentorium (Kernohan’s notch phenomenon).
Additionally, there is a new stroke in the posterior cerebral artery (PCA) territory (light arrow). The
PCA had been compressed by transtentorial herniation
11
CRANIAL NERVE TESTING IN ACUTE NEUROLOGY
Catherine S. W. Albin and Sahar F. Zafar
ANISOCORIA
Step 1: Examine the eyes both in the light and in the dark
Step 2:
THE PUPIL IN LIGHT VS. DARK CAUSES
Difference greater in the dark = Disruption to the sympathetic pathway
MIOSIS First-order neurons: Injury to brainstem and cervical spine, such as
There is a dilation lag meaning in Lateral Medullary Syndrome
the smaller pupil is the Second-order neurons: Pancoast tumor, chest pathology, brachial
abnormal one plexus pathology
Third-order neurons (no associated anhidrosis): internal carotid
artery dissection, neck surgery, cavernous sinus pathology
Difference greater in the light = Disruption to the parasympathetic pathway
MYDRIASIS Cranial nerve III palsy: posterior communicating artery aneurysm,
There is inability of the dilated tumor, temporal lobe uncal herniation
pupil to constrict appropriately Cavernous sinus pathology
Remember that anti-cholinergic drugs can also result in mydriasis.
In hospitalized patients, always look for a scopolamine patch or
recent administration of nebulized ipratropium (in DuoNeb®) to
assess whether the dilated pupil resulted from a medication effect
□ If miosis: look for a Horner’s syndrome (anhidrosis and ptosis), for evidence of a
lateral medullary syndrome (see “Brainstem Syndromes”), or for evidence of
cavernous sinus pathology (testing extraocular eye movement and sensation in
the V1/V2 distribution).
□ If mydriasis: look for disorders of consciousness and consider a STAT scan; if
awake, examine extraocular eye movements and facial sensation, consider CT
angiogram to evaluate for expanding posterior communicating artery aneurysm.
CNIV (Trochlear) Impaired intorsion and depression in the Dorsal midbrain, pineal mass, pathology at The diplopia worsens with head tilted
Nerve Palsy adducted position (superior oblique) the SOF/CVS toward the side of the lesion
14
LESION PRESENTATION COMMON AREAS OF INJURY BEDSIDE TRICKS
CN VI (Abducens) Impaired abduction (lateral rectus) Increased intracranial pressure, cavernous Diplopia is worse with far vision
Nerve Palsy sinus pathology, trauma
Right CN VI palsy:
Medial Longitudinal Results in an internuclear ophthalmoplegia Most commonly multiple sclerosis, or any A right MLF lesion results in impaired
Fasciculus (MLF) Injury (INO): impairs the coordination of stroke/lesion affecting the MLF right eye adduction when looking left
ipsilateral CN III (impaired ipsilateral
adduction) on contralateral gaze Bilateral INO would result in inability
to adduct either eye on horizontal
Right INO: gaze
On left gaze, the right eye does not
adduct and the left eye usually displays
nystagmus
Difference with CN III there is no ptosis
and convergence is not impaired
Patient told “look left”
Nystagmus
15
LESION PRESENTATION COMMON AREAS OF INJURY BEDSIDE TRICKS
One-and-a-half A lesion affects the crossed MLF + PPRF Lesion in the caudal pons This leaves only one horizontal
Syndrome and/or CN VI movement: abduction of the eye which
is contralateral to the lesion
There is no horizontal gaze to the affected
side because of the CN VI/PPRF Often will cause an ipsilateral LMN
involvement pattern of facial weakness (by affecting
CN VII, which is sometimes termed an
Adduction of the ipsilateral eye on
“eight-and-a-half syndrome” (i.e., 7 +
contralateral gaze is impaired due to MLF
1.5)
disruption between CN VI and CN III
Cavernous Sinus (CS) When severe, results in complete Cavernous sinus thrombosis, carotid-
Pathology ophthalmoplegia on the effected side cavernous fistula, pituitary tumor, Tolosa- ICA
Cavernous sinus
Hunt syndrome, pituitary apoplexy Pituitary
Skew Deviation Usually caused by a central lesion Can be caused by lesion to brainstem, Usually the eye on the side of the
16
resulting in vertical misalignment cerebellum or rarely CN VIII injury lesion is higher and intorted
CALORIC TESTING
Both warm and cold water can be used to activate the endolymph of the inner ear
resulting in a current that activates the hair cells. This movement of the hair cells
results in polarization (warm) or hyperpolarization (cold) of the ipsilateral vestibular
verve and apparatus of the brainstem [1]. In the ICU, cold water is preferen-
tially used.
Cold water irrigation of the external auditory canal results in movement of the endo-
lymph in a way that causes hyperpolarization resulting in the inhibition of the vestibu-
lar nerve.
The normal response to cold water:
• A slow movement of the eyes towards from the stimulates, with the fast compo-
nent of nystagmus beating away from the stimulated ear.
In coma:
• There is no corrective saccade because the frontal eye fields are not activated
(due to absent cortical function), thus the eyes will only have the slow movement
towards the cold stimulus.
In brain death testing:
• There is no movement of the eye when the patient’s ear canal is irrigated with
cold water.
17
GENERAL PATTERNS OF FACIAL WEAKNESS
Note that testing facial weakness in less acute patients should involve assessing audi-
tory function (for hyperacusis) and taste. Note that the facial nerve also receives pro-
jections from the extrapyramidal systems and frontal lobe which control emotional
expression. Thus, patients with upper motor pattern of weakness may actually be able
to activate their face involuntarily when associated with an emotional expression.
In comatose patients, the seventh and fifth cranial nerves are assessed by
testing the corneal reflex:
The cornea is the clear layer of tissue over the iris. Touching the cornea transmits a
signal to the brainstem via the Trigeminal Nerve (CN V) and the blink motor response
is carried out due to innervation from the Facial Nerve (CN VII). If either of these are
damaged (such as by a stroke or bleed effecting the brainstem) the patient will not
blink to the gentle touch of the cornea with a cotton swab. Often tested at the bed-
side by dropping a drop of a saline flush into each eye, this is less sensitive but does
not risk injury to the cornea. Take care to actually touch the cornea and not just the
sclera (the white of the eye).
18
TESTING THE GAG REFLEX (PHARYNGEAL REFLEX)
Sensation mediated predominantly by the glossopharyngeal nerve (CN IX). Motor
response mediated by the vagus nerve (CN X). In ICU patients, this is best tested by
advancing a tongue depressor around the endotracheal tube and stimulating the
oropharynx.
REFERENCE
1. Gonçalves DU, Felipe L, Lima TM. Interpretation and use of caloric testing. Braz J
Otorhinolaryngol. 2008;74(3):440–6.
19
STROKE AND VASCULAR ANATOMY
Catherine S. W. Albin and Sahar F. Zafar
Optic chiasm
M1
M2
Classic Syndromes
22
BRANCH TERRITORY
ARTERY OF SUPPLIED CLASSIC SYNDROME IMAGE
Middle ICA Cortical M1 occlusion: Contralateral
Cerebral branches: hemiplegia (including leg because
Artery cortex of of posterior limb of internal
(M1) the lateral/ capsule involvement),
inferior hemianesthesia, and homonymous
frontal hemianopsia (lateral geniculate
lobe, nucleus). Eyes are gazing to the
parietal ischemic side
lobe Left side – global aphasia
Deep Right side – hemineglect and
branches: anosognosia; eyelid opening
putamen, apraxia
part of
caudate,
posterior
limb of
internal
capsule,
corona
radiate
MCA – MCA Frontal eye Face/Arm ≫ Leg weakness. Eye
Superior fields, deviation.
Division Broca’s Left side – Expressive vs. global
area (left), aphasia (acutely) that resolves to
motor and an expressive aphasia sub-acutely.
sensory Right side – Sensory neglect
cortex
23
BRANCH TERRITORY
ARTERY OF SUPPLIED CLASSIC SYNDROME IMAGE
Lateral MCA Putamen, Variable dependent on territory
Lenticulo- part of the affected. Usually hemiplegia,
striate caudate, limited aphasia, homonymous
Arteries posterior hemianopia
(Penetrat- limb of int.
ing capsule,
Branches) outer
globus
pallidus,
corona
radiata
ACA
ACA ICA
MCA
MCA
Midbrain Posterior
communicating
Pons artery
Superior Posterior cerebral
cerebellar artery
artery Basilar artery
Basilar AICA
perforator
Cerebellum PICA
Vertebral artery
Anterior spinal
artery
24
POSTERIOR CIRCULATION
BRANCH TERRITORY
ARTERY OF SUPPLIED SYNDROME IMAGE
Vertebral Subclavian Medial Medial Medulla Syndrome –
Artery medulla, Contralateral hemiparesis
posterior sparing the face,
inferior contralateral loss of position
cerebellum sense, ipsilateral paralysis of
the tongue
Commonly results in an
embolus to the PICA territory,
see below.
Posterior Vertebral Lateral Most commonly affects a
inferior (can be medulla, large posterior territory of
cerebellar Basilar as inferior and the cerebellum (see image)
artery PICA/AICA lateral Lateral Medulla Syndrome –
complex) cerebellum Vertigo, contralateral facial
impaired pain/temp,
Horner’s syndrome,
hoarseness/dysphagia,
vertical diplopia, ipsilateral
ataxia, loss of taste
(Can also occur due to
vertebral artery stroke)
Basilar Intersection Proximal/ In general – crossed
Artery of the Mid symptoms signaling
vertebral Portion – brainstem involvement
arteries penetrating Prox/Mid Portion – Locked-in
branches Syndrome, vertical eye
supply the movements often spared
pons
Top of the Top of the Basilar – Coma
Basilar – (resulting from involvement of
Mid brain the reticular activating
system), disorders of ocular
movement, ptosis, variable
plegia (may be absent),
behavioral abnormalities
(akinetic mutism, visual
hallucinations)
25
BRANCH TERRITORY
ARTERY OF SUPPLIED SYNDROME IMAGE
Anterior Basilar Middle Ipsilateral ataxia; nausea/
Inferior cerebellar vomiting/slurred speech.
Cerebellar peduncle; Occasionally may see loss of
Artery inferolat- pain and temp contralateral,
eral pons, ipsilateral Horners, paresis
flocculus, of conjugate lateral gaze or
anteroinfe- tinnitus.
rior surface
of cerebel-
lum
Superior Basilar Middle and Ipsilateral ataxia; nausea/
Cerebellar superior vomiting/slurred speech.
Artery cerebellar Can see loss of pain and
peduncles, temperature contralateral
rostral due to involvement of the
cerebellum spinothalamic tract.
to the
horizontal
fissure,
portion of
midbrain
Posterior Basilar Cerebral Presentation variable given
Cerebral peduncles, this artery supplies the rostral
Artery CN III and brainstem, inferior medial
(PCA) IV, temporal lobes, and
thalamus, thalamus. Strokes affecting
hippocam- the midbrain can cause
pus and palsies of vertical gaze,
medial stupor/coma, or CN III
temporal palsies. Thalamic syndromes
lobe, can mimic any other
occipital syndrome. Occlusion of the
lobe cortical branches result in
homonymous hemianopia,
alexia without agraphia (left
PCA), anomia. Amnesic
syndromes.
26
BRANCH TERRITORY
ARTERY OF SUPPLIED SYNDROME IMAGE
Artery of P1 A variant in Disorder of consciousness –
Percheron which the usually somnolence,
thalamoper- sometimes with associated
forate hemianesthesia or
branches hemiplegia.
arises from
one side of
the P1
segment
and supply
BOTH
medial
thalami
Lacunar Syndromes
SYNDROME LOCATION
Pure Motor Hemiplegia Internal capsule, corona radiata, ventral pons
Pure Sensory Stroke Lateral thalamus or deep parietal white matter
Ataxic Hemiparesis Anterior pons, midbrain at the cerebral peduncle (rare), internal capsule
Clumsy Hand-Dysarthria Paramedian mid-pons contralateral to symptoms, posterior portion of
internal capsule
REFERENCE
1. Ropper AH, et al. Adams and Victor’s principles of neurology. McGraw Hill Medical; 2005.
27
BASICS OF COMPUTED TOMOGRAPHY (CT)
Catherine S. W. Albin and Sahar F. Zafar
While the detail of neuroanatomy is much more sensitive with MRI, non-contrasted
head CTs (NCHCT) have the advantage of being faster, more readily available, more
affordable, and can be used in patients that have implants/hardware that are not MRI
compatible.
Acute hemorrhage is hyperdense (bright white) on CT, which makes it the ideal
screen for hemorrhage. Vasogenic, interstitial, and cytotoxic edema are hypodense,
which can be more difficult to see, but can be detected by looking for symmetry
between the two sides of the brain. Anatomic distortions, like herniation and hydro-
cephalus, are easily detected with CT, which make it ideal for assessing these
findings. Contrast can be added to look for breakdown of the blood-brain barrier.
4 4 4
7
3 3
2 2 3
1 2 1 1
5 5 5
6
6 6
30
AT THE LEVEL OF THE
AT THE LEVEL OF THE CAUDATE THALAMUS NEAR THE VERTEX
5 6
4
7 5
4 4 1
3 3 7
1 1 2
2
2 6
3
31
Acute hemorrhage will appear bright and can be further categorized by the shape of the bleed and
its location.
Blood fills the cisterns and Crescent-shaped collection. Lens-shaped collection. Often
fissures and layers around the Chronic blood will become formed by damage to the middle
parenchyma. hypo/isodense and is harder to meningeal artery or its collateral
detect. supply.
INTRAPARENCHYMAL INTRAVENTRICULAR
HEMORRHAGE HEMORRHAGE
32
FEATURES CONCERNING FOR HYDROCEPHALUS
ENLARGEMENT OF THE BOWING OF THE 3RD ENLARGEMENT OF THE
TEMPORAL HORNS VENTRICLE LATERAL VENTRICLES
33
USE OF COMPUTED TOMOGRAPHY IN STROKE
The primary use of use of CT in stroke is to exclude hemorrhage. However, CT can
also demonstrate early ischemic changes, by which the patient is assigned an
ASPECTS score (see page 67).
An example of early ischemic changes:
Fig. 6.1 Non-contrast Head CT in Acute Ischemic Stroke: The Image demonstrates subtle blurring
of the gray–white differentiation of the cerebral cortex and white matter tracts (marked with a fat
arrow). There is also effacement of the sulci (marked with skinny arrow)
CT Angiogram Head & Neck is also of paramount importance in stroke workup and
management especially in screening for a Large Vessel Occlusion (LVO) which would
make the patient a candidate for thrombectomy. Although reviewing CT angiograms
is beyond the scope of this chapter, a few important tips for reviewing CTAs:
The “CTA Thins” will provide the source images and should be reviewed first.
Practice reviewing each major extracranial to intracranial artery (right and left internal
carotid arteries, right and left vertebral arteries, and basilar artery), and then tracing
the major branches -- middle cerebral, anterior cerebral, and posterior cerebral
arteries. A helpful tip in distinguishing the internal from external carotid arteries after
they branch from the common carotid is that the external carotid has multiple
branches in the neck, whereas the internal carotid’s first branch is the ophthalmic
artery which is after the artery has entered the skull base.
34
Fig. 6.2 CTA Thin, Neck: at the take-off of the left internal carotid from the common carotid and at
the very distal right common carotid. There is some calcified plaque at the distal right common
carotid (hyperdense, marked with thin arrows). Additionally, there is non-calcified plaque in the
proximal left internal carotid causing significant stenosis in the left take off. The lumen is marked
with a circle and the soft hypodense plaque is marked with a fat arrow). This soft plaque likely
resulted in an embolic stroke to the left MCA territory a so-called artery-to-artery stroke—i.e. the plaque
came from a larger artery and embolized to a distal intracranial artery
35
The “MIP” (maximum intensity projection) reformatting of blood vessels make it
easier to visualize the major intracranial vessels, and these sequences are the
easiest to review when attempting to detect large vessel occlusions.
Fig. 6.3 Axial MIP in Acute Stroke: Image demonstrates complete lack of opacification of the right
intracranial internal carotid artery and middle cerebral artery in a patient with acute thrombus at the
ICA Terminus
36
BASICS OF MAGNETIC RESONANCE IMAGING (MRI)
ORDERING AND ASSESSMENT
Catherine S. W. Albin and Sahar F. Zafar
MRI SEQUENCES
HYPERINTENSITY VS.
SEQUENCE HYPOINTENSITY BEST FOR: EXAMPLE
T1 Sequences White matter is light gray • Anatomy
(spin echo): Gray matter is dark gray • Post-contrasted
MPRAGE T1 Bright: images
images are T1 • Gadolinium contrast (gadolinium is
weighted and • Fat not apparent on
very high • Subacute hemorrhage T2 weighted
special • Protein-rich fluid sequences)
resolution • Early subacute blood
T1 Dark: T1 Post-Gadolinium:
• CSF Heterogeneous cavity
• Inflammation, edema, enhancement seen in a
demyelination patient with a high-grade
• Chronic blood products glioma.
T2 Sequence White matter is dark gray • Anatomy
(spin echo): Gray matter is light gray • Chronic
FLAIR is a T2 T2 Bright: pathology
weighted • Demyelination, axonal loss • Edema
image • Slow-flow through blood vessels • Demyelination
• Cytotoxic and vasogenic
edema
• Subacute to chronic infarcts
• Hyperacute and late subacute T2 FLAIR: Posterior white
• Air Encephalopathy
38
MRI IN STROKE
SEQUENCE REVIEW FOR: EXAMPLE
DWI Ischemic core (DWI Bright)
39
SEQUENCE REVIEW FOR: EXAMPLE
GRE/SWI • Hemorrhagic transformation/
petechial (SWI/GRE dark)
• Chronic microbleeds, may
identify chronic hypertension
and/or signature of
possible/probable CAA
• Clot in the artery (identified
by a “blooming artifact”
within the affected vessel)
40
MRI IN HEMORRHAGE
SWI/GRE These sequences can demonstrate
hemorrhage; SWI imaging is the
most sensitive for detecting small
amounts of blood and can be used
to detect microhemorrhages and
help differentiate between CAA and
hypertensive hemorrhages.
41
MRI IN INFECTIOUS/INFLAMMATORY/NEOPLASTIC CONDITIONS
T1 As gadolinium can demonstrate active
w/gadolinium areas of blood brain barrier breakdown,
T1 with gadolinium should always be
obtained in cases suspected of
demyelination (such as MS, ADEM,
hereditary demyelinating conditions),
neoplastic conditions (gliomas,
lymphomas, menigiomas, etc.), and
encephalitis/meningitis. These images
also demonstrate inflammation of T1 Post-Gadolinium: Contrast-enhancing
meninges such as seen in meningitis and meningioma in the extra-axial space
leptomeningeal carcinomatosis, and may causing compression and midline shift
reveal characteristic “ring enhancement” of the left frontotemporal lobe.
patterns that help narrow the differential
diagnosis. For examples of T1 post-
gadolinium patterns, see below.
T2 FLAIR Essential in determining the pattern of
inflammation and thus providing a major
diagnostic clue to the etiology.
Encephalitis patterns may be more
specifically described as limbic,
rhomboencephalitic, cerebellitis, and
basal ganglia inflammation.
42
MRI IN INFECTIOUS/INFLAMMATORY/NEOPLASTIC CONDITIONS
MR Perfusion Advanced imaging technique that
measures the degree of angiogenesis and
capillary permeability, it is particularly
helpful in grading gliomas and can
provide prognostic information.
43
EXAMPLES OF DIFFUSION RESTRICTION PATTERNS
ACUTE ISCHEMIA HYPOXIC-ISCHEMIC DAMAGE ABSCESS
44
EXAMPLES OF CONTRAST ENHANCEMENT
RING-ENHANCEMENT VENTRICULITIS MENINGITIS
REFERENCE
1. Mandell DM, et al. Intracranial vessel wall MRI: principles and expert consensus recommenda-
tions of the American Society of Neuroradiology. Am J Neuroradiol. 2017;38(2):218–29.
45
UNDERSTANDING TRANSCRANIAL DOPPLERS (TCDS)
Catherine S. W. Albin and Sahar F. Zafar
60
-60
60
-60
48
MONITORING
• There are four standard TCD “acoustic windows.” The transtemporal window (for
the MCA, ACA, and PCA), transorbital (carotid siphon and ophthalmic artery),
suboccipital window (basilar and vertebral arteries), and sometimes, the subman-
dibular window (distal ICA)
• The different branches of the Circle of Willis are identified by the speed of flow,
direction of flow, and a characteristic depth at which the artery should be found.
APPLICATION IN SAH
• TCDs can be used to help evaluate and diagnose a patient with vasospasm. In
studies, moderately elevated mean velocities (120–199 cm/s) in the MCA did not
always correlate with angiographic spasm; however, the positive predictive value of
mean velocities ≥200 cm/s was 87%. The negative predictive value for mean
velocities <120 cm/s was 94%. Thus, very elevated or low cerebral artery flow
velocities reliably predict angiographically significant vasospasm or the lack
thereof [2].
• The Lindegaard Ratio (LR) is an important ratio in evaluating for vasospasm:
Since velocities can also be influenced by hematocrit, blood pressure, and CO2,
etc., the Lindegaard Ratio accounts for the difference in flow velocity in the ICA and
MCA. The MCA and ICA should be equally affected by patient factors, but the ICA
should not be affected by vasospasm.
–– A Lindegaard Ratio (LR) of <3 is normal. An LR of 3–6 is suggestive of mild-
moderate spasm.
–– An LR >6 is considered to be indicative of severe spasm.
• Institutions use slightly different cut offs for acceptable upper limits of normal;
however, a general guide is:
• It is also important to take into account the degree of change and the amount of
time that change occurred over. For example, a 20% increase in velocity in one
day, even if not reaching a threshold of LR >3, should still raise concern.
• TCD reports will also include the pulsatility index (PI). The pulsality index reflects
the change between systolic and diastolic pressures. An increase in the pulsatility
index may represent downstream resistance to flow. This is often as a result of
distal vasospasm or increased intracranial pressure. An abrupt rise in the PI or a
PI>1.5 should alert you to these possibilities.
49
OVERVIEW OF OTHER TCD APPLICATIONS
• In ischemic stroke: TCDs can be used to track arterial occlusion before and after
tPA [4]. TCDs with emboli detection (HITS – high-intensity transient signals) can be
used to assess the risk for ongoing thrombo-embolic disease and may be helpful in
determining the need for aggressive therapy (such as anticoagulation) [5].
• Brain death testing: As ICP rises, CPP approaches zero leading to cerebral
circulatory arrest. When ICP is equal to the diastolic perfusion pressure, the dia-
stolic perfusion drops to 0 and there is no diastolic flow seen on TCDs. As the ICP
continues to rise, the diastolic flow reverses resulting in net forward flow of zero,
which is consistent with cerebral circulatory arrest [6].
Fig. 8.3 The above Transcranial Doppler display demonstrates oscillating flow and thus cessation of
cerebral perfusion. There is forward flow, but ICP is equal or greater than SBP, and thus during dias-
tole there is flow reversal of the same magnitude and the net forward flow is 0. This is consistent with
cerebral circulatory arrest
REFERENCES
1. Purkayastha S, Sorond F. Transcranial doppler ultrasound: technique and application. In:
Seminars in neurology, vol. 32. Thieme Medical Publishers; 2012.
2. Vora YY, et al. Role of transcranial Doppler monitoring in the diagnosis of cerebral vasospasm
after subarachnoid hemorrhage. Neurosurgery. 1999;44(6):1237–48.
3. Sviri GE, et al. Transcranial Doppler grading criteria for basilar artery vasospasm. Neurosurgery.
2006;59(2):360–6.
4. Christoe I, et al. Timing of recanalization after tissue plasminogen activator therapy deter-
mined by transcranial doppler correlates with clinical recovery from ischemic stroke. Stroke.
2000;31(8):1812–6.
50
5. Daffertshoker M, et al. High-intensity transient signals in patients with cerebral ischemia. Stroke.
1996;27(10):1844–9.
6. Ducrocq X, Hassler W, Moritake K. Consensus opinion on diagnosis of cerebral circulatory arrest
using Doppler-sonography: Task Force Group on cerebral death of the Neurosonology Research
Group of the World Federation of Neurology. J Neurol Sci. 1998;159:145–50.
51
TIPS AND TRICKS FOR EEG INTERPRETATION
Catherine S. W. Albin and Sahar F. Zafar
Please note a comprehensive review of EEG is outside of this guide’s scope. Below
is an abbreviated overview.
Frequencies (named for the order in which they were discovered)
Large peaks counted between 1 sec intervals
Beta >13 hz Seen normally in the frontocentral leads when awake. In the ICU, often a result of
sedation (benzodiazepines classically, propofol)
Alpha 8–13 hz Normal background activity best seen in the posterior leads when a healthy
patient’s eyes are closed
Theta 5–7 hz Slower frequency that can be associated with normal drowsiness or
encephalopathy
Delta <5 hz Seen normally in sleep. In the ICU, associated with encephalopathy and structural
lesions (focal delta)
Fig. 9.1 EEG demonstrating 3Hz spike-and-wave discharges consistent with seizure
Note that sometimes an EEG may be noted as becoming “faster,” which can mean
one of two things:
1. The BACKGROUND may be faster, such as moving from mainly delta to theta, for
example. This is a good thing as generally this suggests the patient is waking up.
54
ELECTRODES
Even electrodes are on the right, odd electrodes are on the left
Fp1 Fp2
F7 F8
F3 F4
Fz
A1 T3 C3 Cz C4 T4 A2
P3 Pz
P4
T5 T6
O1 O2
• EEG can be viewed in different montages. A “Double Banana” montage is one that
is commonly used as it allows for easy comparison between the left and right
hemispheres.
• Set the sensitivity (how “loud” the data is): the default is 7 uV/mm, you can use the
keyboards “up” and “down” arrows to change this (for example, 15 uV/mm will
make the amplitude lower)
• Scroll through by either arrowing “left” and “right” or by hitting the space bar to play
the data. Note that you can find out the exact time by looking at the very top of
the screen.
• The HFF (high frequency filter) can “clean up” data and is a way to reduce muscle
artifact. The brain does not generate frequencies much faster than 15hz. This
allows you to “filter out” any data that is faster than whatever this is set to.
However, note that by minimizing artifact, you are also compromising the data and
may also reduce “signal” while reducing “noise”
55
GETTING THE BIG PICTURE (THE SPECTROGRAM AND SEIZURE DETECTION)
The spectrogram is a way to help comb through hours and hours of data by giving a
power analysis. At least a snippet of the EEG should be reviewed for each part of the
spectrogram that appears different.
What Is It:
• The spectrogram is a computer algorithm (the details of which are beyond the
scope of this chapter) that processes the sinusoids of the EEG and represents the
amplitude of the sinusoids as a function of frequency
• The x-axis=time; the y=frequency, color corresponds to power
• The seizure detection feature is at the bottom of this toolbar. It does not replace
review of raw EEG data. However, it can be used to pinpoint time to review
the data.
How to Use It:
• Using the Spectrogram:
–– The window of time to view can be adjusted (see image, next page)
–– How does the pattern change when sedation is held? Or when are the
AEDs loaded?
–– Are there any periods of sharp changes?
• Using the Seizure Probability feature
–– Click a red “spike” to take you to this time and scroll a bit before. What does the
background look like? How does it change as you move towards the
“Seizure” spike?
–– What is the patient doing? Check by the video camera!
Tips for Reading EEG:
• Given limited time in pre-rounding, select sections of the EEG can be reviewed
based on clinical changes, bedside nursing annotation, medication administration,
or changes seen in the spectrogram. Note: this does not replace a detailed review
of all EEG data and should be performed per institutional practice
• Assess the overall organization: What is the dominant background frequency -
Theta? Delta? Is a Posterior Dominant Rhythm (alpha activity in the posterior leads)
present?
• Assess the symmetry, what is the predominant frequency on the right side /
left side?
• Are there any periods of burst suppression?
• Are there any sharp waves, periodic discharges?
• Seizures? And if so, how many?
• Page 267 reviews some of the findings on the inter-ictal continuum and discusses
an algorithm for their management.
56
Fig. 9.4 A Spectrogram
57
PART II
VASCULAR NEUROLOGY
ACUTE ISCHEMIC STROKE – FIRST ENCOUNTER ASSESSMENT
AND MANAGEMENT
Catherine S. W. Albin and Sahar F. Zafar
For eligible patients, the goal door to needle time for IV tPA/TNK is <60 min (Class I
Evidence) [5]
Multiple tasks should be performed by the ED team (physician, advanced practice
provider/resident, radiologist, CT tech, pharmacy, registrar, and nursing):
• EMT/ED teams flag possible acute stroke and notify neurology and radiology teams
• Patient is registered and assigned a medical record number
• ED team assesses ABCs and stabilizes patient
• Radiology is contacted about need for STAT CT/CTA/CTP (imaging protocols vary
by institution).
• Screening labs and a POCT glucose are obtained. Labs should be sent STAT.
• At least an 18G IV is placed for administration of contrast
• Ideally a travel bedside monitor is set up so that the patient can be immediately
transferred to the CT scanner
• Pharmacy is alerted about potential need for tPA and someone with experience
mixing tPA is available/ready to mix tPA if needed Mixing is not required for TNK.
In addition to working with the ED team to ensure the above tasks are being carried
out, the neurologist’s role is to:
□□Arrive to assess the patient as soon as possible
□□Confirm that the patient is ordered for a noncontrast head CT and standard
institutional protocol for additional imaging
□□Establish time of onset/last seen well (LSW)
□□Document National Institute of Health Stroke Score (NIHSS)
© The Author(s), under exclusive license to Springer Nature Switzerland AG 2022
C. S. W. Albin, S. F. Zafar (eds.), The Acute Neurology Survival Guide,
https://doi.org/10.1007/978-3-030-75732-8_10
61
□□Investigate if the patient is on anticoagulation, and if it cannot be determined
but there is a high likelihood based on the patient’s known medical history,
confirm that coags, including anti-Xa assay for novel oral anticoagulants (if
available), are sent.
□□ Review patient’s history for contraindications for IV tPA/TNK (see next page),
□□ Discuss risks/benefits of tPA/TNK and other treatment options with family/
patient (see next page). Note that it may be required to have documentation of
consent in the chart.
□□ Review if patient is an endovascular therapy candidate (see page 67). If CTA is
not readily available, do not delay tPA/TNK administration for vessel imaging.
If the Patient has no hemorrhage and is within 4.5 h of last seen well and meets
other criteria for TPA:
□□ Weigh patient
□□ Confirm (again!) the patient’s blood pressure is <185/110, if not, use labetalol,
nicardipine, or clevidipine to lower.
□□ Dosing tPA: 0.9 mg/kg IV; not to exceed 90 mg total dose; administer 10% of
the total dose as an initial IV bolus over 1 min; remainder is infused
over 60 min
□□ Dosing TNK: There are variable dosing strategies. 0.25 mg/kg (max 25mg) was
used in NEJM trial [3] administered over 5 seconds
□□ Once tPA/TNK is administered, let BP autoregulate no higher than 180/105 for
24 h post-tPA. Blood pressure should be monitored every 15 min for the first
24 h, then every 30 min for 6 h, and then every 60 min until 24 h after
treatment.
62
□□Current use of anticoagulant with INR >1.7 or PT >15 s
□□Treatment doses of a low molecular weight heparin (ie enoxaparin) within 24 h,
or current use of direct thrombin inhibitors or factor Xa inhibitors, if last dose
within 48 h
□□ Extensive hypodensity on CT, suggesting that the new infarct has already
completed. Prior strokes (> 3 months prior) do not exclude the patient
from tPA/TNK.
Relative Contraindications (From ASA/AHA Guidelines [5])
□□ Only minor or rapidly improving stroke symptoms
□□ Seizure at onset with post-ictal residual neurological impairments
□□ Major surgery or serious trauma (within previous 14 days)
□□ Intracranial vascular abnormalities that do not meet threshold for absolute
contraindication per AHA 2018 guidelines
□□ Pregnancy
□□ Blood glucose <50 or >400 mg/dL
Other Considerations if Dosing TPA in the Extended Window (3–4.5 h)
The original ECASS III trial [6] for extended window tPA excluded patients >80 years,
patients on anticoagulation, and patients with a history of stroke and diabetes;
however, follow up data [5] has demonstrated:
□□ Patient >80 y.o. also benefit from tPA in extended window (Class IIA Evidence)
□□ In patients with prior stroke and diabetes, extended window tPA may be
reasonable (Class IIB evidence)
□□ If patient is taking warfarin but INR ≤1.7, tPA appears safe and may be benefi-
cial (Class IIB)
□□ The benefit of IV tPA for patients with a very high NIHSS (>25) is unknown
(Class IIB)
63
For Extended Window (3–4.5 h) (based on ECASS III Data [6]):
• tPA administered in this window is associated with a modest improvement in
favorable outcomes. Of patients treated with tPA in this timeframe about 7 of
100 patients will have a better outcome than if they did not receive tPA.
• Patients that receive tPA in this time window have a higher rate of brain bleed-
ing, but only about 3 patients of every 100 patients treated have a symptom-
atic bleed.
• The FDA has not approved tPA to be given after 3 h, but we have trial data that
suggest it is still safe and efficacious.
64
For thrombectomy capable centers assessing patients either in the ED or for
transfer: Both clinical and radiographic data determine how likely the patient is to
benefit from thrombectomy:
The use of advanced neuroimaging (most often CTP) should be done based on hospital protocols
*Note that a recent retrospective analysis showed that with recanalization even patients with
ASPECTS <5 could achieve good mRS scores at 90 days and were nearly 5 times more likely to
achieve favorable outcome [20].
As such, discussion with an endovascular capable center is advised even in patients with a less
favorable ASPECT score. Large-core randomized trials are currently underway to further guide this
management.
65
TPA+MECHANICAL THROMBECTOMY (MT) VS. ONLY MT
Three trials (DEVT [15], DIRECT-MT [16], SKIP [17]) came out in early 2021 that
looked for non-inferiority of mechanical thrombectomy (MT) alone when compared to
IV tPA+MT (tPA+MT). This is referred to as “bridging” with tPA.
Although two of these trials demonstrated non-inferiority of bypassing IV tPA there
was also major criticism that the non-inferiority margins were overly generous, and
that SKIP was underpowered.
There is nuance and local preference but some factors that may favor MT alone
include situations in which peri-procedural dual-antiplatet therapy may be needed:
such as for tandem ICA/MCA occlusion, dissections, and significant intracranial
atherosclerosis, or when the MT team is rapidly available. IV tPA may offer higher
benefit to patients with very early presentations, long travel time to MT capable
center, or when there is a borderline low NIHSS for thrombectomy [18].
At the time of this publication, IV tPA/TNK is still the standard of care for all
patients presenting within the appropriate window whether or not MT is
being pursed.
66
MAJOR SCORING METRICS FOR ACUTE STROKE
ASPECTS SCORING
Calculating an Alberta Stroke Program Early CT Score (ASPECTS) [6]
Each of the 10 areas shown below is assigned a point.
Whenever there is blurring of the gray–white differentiation of an area (suggestive of
early ischemic change) then a point is subtracted for that area.
A score of 10 is a normal head CT.
A score of 0 suggests radiographically detectable ischemic change in the entire MCA
territory.
Fig. 10.1 Example of ASPECTS Territories. C = Caudate, L = Lentiform Nucleus, IC = Internal capsule
67
REFERENCES
1. Burgos AM, Saver JL. Evidence that tenecteplase is noninferior to alteplase for acute ischemic
stroke: meta‐analysis of 5 randomized trials. Stroke. 2019; 50:2156–62.
2. Campbell BC, Mitchell PJ, Churilov L, Yassi N, Kleinig TJ, Yan B, Dowling RJ, Bush SJ, Dewey
HM, Thijs V, et al. Tenecteplase versus alteplase before endovascular thrombectomy (EXTEND‐
IA TNK): a multicenter, randomized, controlled study. Int J Stroke. 2018; 13:328–34.
3. Campbell, Bruce CV, et al. Tenecteplase versus alteplase before thrombectomy for ischemic
stroke. N Eng J Med 2018;378(17):1573-82.
4. National Institute of Neurological Disorders and Stroke rt-PAStroke Study Group. Tissue plas-
minogen activator for acute ischemic stroke. N Engl J Med. 1995;333:1581–7.
5. Powers WJ, et al. 2018 guidelines for the early management of patients with acute ischemic
stroke: a guideline for healthcare professionals from the American Heart Association/American
Stroke Association. Stroke. 2018;49(3):e46–99.
6. Hacke W, et al. Thrombolysis with alteplase 3 to 4.5 hours after acute ischemic stroke. N Engl J
Med. 2008;359(13):1317–29.
7. Jovin TG, Chamorro A, Cobo E, et al. Thrombectomy within 8 hours after symptom onset in isch-
emic stroke. N Engl J Med. 2015;372(24):2296–306. https://doi.org/10.1056/NEJMoa1503780.
8. Saver JL, Goyal M, Bonafe A, et al. Stent-retriever thrombectomy after intravenous t-PA vs. T-PA
alone in stroke. N Engl J Med. 2015;372(24):2285–95. https://doi.org/10.1056/NEJMoa1415061.
9. Campbell BC, Mitchell PJ, Kleinig TJ, et al. Endovascular therapy for ischemic stroke with
perfusion-imaging selection. N Engl J Med. 2015;372(11):1009–18. https://doi.org/10.1056/
NEJMoa1414792.
10. Goyal M, Demchuk AM, Menon BK, et al. Randomized assessment of rapid endovascular
treatment of ischemic stroke. N Engl J Med. 2015;372(11):1019–30. https://doi.org/10.1056/
NEJMoa1414905.
11. Nogueira RG, Jadhav AP, Haussen DC, et al. Thrombectomy 6 to 24 hours after stroke with a
mismatch between deficit and infarct. N Engl J Med. 2018;378(1):11–21. https://doi.org/10.1056/
NEJMoa1706442.
12. Albers GW, et al. Thrombectomy for stroke at 6 to 16 hours with selection by perfusion imaging.
N Engl J Med. 2018;378(8):708–18.
13. Pexman JHW, et al. Use of the Alberta Stroke Program Early CT Score (ASPECTS) for assessing
CT scans in patients with acute stroke. Am J Neuroradiol. 2001;22(8):1534–42.
14. Thon, Jesse M., and Tudor G. Jovin. “Imaging as a selection tool for thrombectomy in acute isch-
emic stroke: pathophysiologic considerations.” Neurology 97.20 Supplement 2 (2021): S52-S59.
15. Zi W, et al. Effect of endovascular treatment alone vs intravenous alteplase plus endovascular
treatment on functional independence in patients with acute ischemic stroke: the DEVT random-
ized clinical trial. JAMA. 2021;325(3):234–43.
16. Yang P, Zhang Y, Zhang L, Zhang Y, Treurniet KM, Chen W, Peng Y, Han H, Wang J, Wang
S, DIRECT-MT Investigators, et al. Endovascular thrombectomy with or without intravenous
alteplase in acute stroke. N Engl J Med. 2020;382:1981–93.
68
17. Suzuki K, et al. The randomized study of endovascular therapy with versus without intravenous
tissue plasminogen activator in acute stroke with ICA and M1 occlusion (SKIP study). Int J Stroke.
2019;14(7):752–5.
18. Nogueira RG, Tsivgoulis G. Large vessel occlusion strokes after the DIRECT-MT and SKIP trials:
is the alteplase syringe half empty or half full? Stroke. 2020;51(10):3182–6.
19. Writing Group for the BASILAR Group. Assessment of endovascular treatment for acute basilar
artery occlusion via a nationwide prospective registry. JAMA Neurol. 2020;77(5):561–73.
20. Almallouhi E, Al Kasab S, Hubbard Z, et al. Outcomes of Mechanical Thrombectomy for Patients
With Stroke Presenting With Low Alberta Stroke Program Early Computed Tomography Score in
the Early and Extended Window. JAMA Netw Open. 2021;4(12):e2137708.
69
PERFUSION IMAGING
Catherine S. W. Albin and Sahar F. Zafar
The goal of perfusion imaging is to determine the extent of territory at risk of infarction
so as to correctly triage the patients that are most likely to benefit from endovascular
reperfusion therapies [2].
Terms
Cerebral Blood Volume (CBV): Total cerebral blood volume in a given unit of
brain volume (mL/100 g)
Cerebral Blood Flow (CBF): Total volume of blood moving through a given unit of
brain volume per unit time (mL/100 g/min)
Mean Transit Time (MTT): Average transit time of blood through a given brain
region in seconds
Perfusion–Diffusion Mismatch: Finding a difference in the tissue that is ischemic
and the tissue that is at risk of infarction (the penumbra)
TECHNIQUES [1]
• CT Perfusion: Uses iodinated contrast to generate MTT, CBV, and CBF measure-
ments. Main benefit is that it can be rapidly obtained. Major limitations are that it is
less sensitive in detecting the ischemic core as MRI and exposes patients to
contrast.
• MR Perfusion: Uses gadolinium contrast agent to trace the perfusion of blood and
generate the above measurements. The three types are described below.
–– Dynamic Susceptibility Contrast (DSC) MR Perfusion: registers the susceptibility-
induced signal loss on T2-weighted sequences after a bolus of gadolinium-based
contrast passes through a capillary bed.
–– Dynamic Contrast-Enhanced (DCE) MR Perfusion: measures T1 shortening due
to gadolinium-based contrast passing through tissue.
–– Arterial Spin Labeling (ASL): Water molecules are magnetically “labeled” by
selective radiofrequency (RF) irradiation pulse applied to the neck, and then a
downstream measurement of the labeled water molecules are collected in the
brain. This generates a map of cerebral blood flow. Benefits include that it requires
no contrast agent. Major limitation is that there is a low signal-to-noise ratio which
can make the data difficult to interpret.
72
EXAMPLE OF IMAGING IN A PATIENT EXAMPLE OF IMAGING IN A PATIENT THAT
THAT HAS ALREADY ESTABLISHED AND HAS A SMALL ISCHEMIC CORE AND A
ISCHEMIC CORE: LARGE PENUMBRA:
CBF Markedly decreased Mildly decreased
REFERENCES
1. Campbell BC, Christensen S, Levi CR, et al. Comparison of computed tomography perfusion
and magnetic resonance imaging perfusion-diffusion mismatch in ischemic stroke. Stroke.
2012;43(10):2648–53.
2. Menon BK. Neuroimaging in acute stroke. Continuum. 2020;26(2):287–309.
73
ISCHEMIC STROKE: ADMISSION CHECKLIST
Catherine S. W. Albin and Sahar F. Zafar
REVIEW
Note class of evidence based on the ASA/AHA 2018 Acute Ischemic Stroke
Guidelines [1]
□□Review non-contrasted head CT (NCHCT), CTA head & neck, and perfusion
imaging (as available)
□□Confirm the patient received tPA/TNK (Class I), if no contraindications (see
page 63) or received ASA 325 mg, if not eligible for tPA/TNK and no contraindi-
cation for aspirin. If the patient received tPA/TNK, the first dose of aspirin
should be held until a 24 hour head CT demonstrates no significant intracranial
hemorrhage
□□If s/p thrombectomy, review imaging and receive sign-out from endovascular
team, clarify blood pressre goals. Note: Recent trial comparing SBP goals in
thrombectomy found no difference in outcomes for patient with an avg BP of
128 mmHg (intensive BP loweing group) vs 138 mmHg (control group [3]).
□□EKG (Class I)
SPECIAL CONSIDERATIONS
□□If s/p tPA: no foley unless necessary to prevent obstructive acute kidney injury
□□if s/p tPA: 24-h head CT to confirm no hemorrhagic transformation
□□If severe carotid stenosis or extracranial dissection, consider anticoagulaton
(Class IIb). See Symptomatic Carotid Stenosis, page 95 and Dissection,
page 89
□□ Consider if early hemicraniectomy is warranted for malignant cerebral edema
(see page 199) and consider the need for hyperosmolar therapy (see
page 195).
□□ Consider the age, stroke risk factors, and comorbidities of a patient before
sending additional lab work (see page 79)
76
Notes for Hypercoagulation Testing:
Warfarin Affects:
Protein C & S testing
REFERENCES
1. Powers WJ, et al. Guidelines for the early management of patients with acute ischemic stroke:
2019 update to the 2018 guidelines for the early management of acute ischemic stroke: a
guideline for healthcare professionals from the American Heart Association/American Stroke
Association. Stroke. 2019;50(12):e344–418.
2. Johnston SC, et al. Platelet-oriented inhibition in new TIA and minor ischemic stroke (POINT)
trial: rationale and design. Int J Stroke. 2013;8(6):479–83.
3. Mazighi, M et al. Safety and efficacy if intensive blood pressure lowering after successful endo-
vascular therapy in acute ischaemic stroke (BP_TARGET): a multicentre open-label, randomized
controlled trial. The Lancet Neurology 2021;20(4):265–74.
77
STROKE WORKUP – BEYOND THE BASICS
Catherine S. W. Albin and Sahar F. Zafar
80
ISCHEMIA DUE TO VENOUS HYPERCOAGABLE STATE (REQUIRES R → L SHUNT)
STATE- SYSTEMIC
DEPENDENT DISEASES
INHERITED (I.E. (INCLUDING BUT
THROMBOPHILIAS [7] PROVOKED) DRUGS NOT LIMITED TO)
Etiology Factor V Leiden mutation, Trauma, Oral contraceptive Peripheral noctural
prothrombin gene pregnancy, pills, hormonal hemoglobinuria
mutation, protein C/S surgery, therapy, (PNH)
deficiency, antithrombin III prolonged testosterone, Nephrotic syndrome
deficiency immobility lupus-inducing Inflammatory bowel
medications, disease
bevacizumab
Clues Personal or family history Evident by Appropriate Often evident by
of venous thrombosis history medication list history or comorbid
findings.
Lab Tests for above etiologies, None needed None needed Beyond the scope of
evaluation often available as a this chapter
hypercoaguable panel
(see page 77 for how
anticoagulation and acute
disease effects this testing)
Imaging Deep vein thrombosis DVT screen DVT screen DVT screen
evaluation (DVT) screen
Notes: Factor V Leiden mutation
is 5–10× more common
than the others.
Acquired causes of
protein C/S and anti-
thrombin deficiency (DIC,
acute thrombosis,
cirrhosis, nephrotic
syndrome, ECMO, drug
interaction) are much
more common than
inherited causes
81
ISCHEMIA DUE TO LESS COMMON CAUSES OF THROMBOEMBOLIC DISEASE [8]
INFECTIVE MARANTIC PARADOXICAL
ENDOCARDITIS ENDOCARDITIS DISSECTION EMBOLUS
Etiology Most common Deposition of sterile Injury to great vessels Venous clot – assess
pathogens: thrombi often due to either by trauma or for the risk factor
• Staphylococcus endothelial injury genetic connective described above
(MSSA and from a tissue disorder (many that traverses a
MSRA) hypercoagulable have probable right-to-left shunt
• Streptococcus state. association, but such as a PFO or
(viridians and Often seen with SLE fibromuscular ASD
bovis) and adenocarcinoma dysplasia (FMD) is
• Enterococcus likely the most
Culture negative common)
pathogens:
• HACEK
organisms
• Coxiella
• Bartonella
• Tropheryma
whippelii
Clues Fevers, chills, night
Depends on etiology, Horner’s syndrome if DVT, risk factors for
sweats + new may have no carotid involved venous
murmur associated symptoms Neck trauma by hypercoagable
history, or neck pain state (as per above)
or posterior orbital
pain/headache
Lab Blood cultures, ESR/CRP, screen for Per above section
evaluation ESR/CRP malignancy and
rheumatologic
conditions
Imaging TTE and if negative TTE and if negative MRA with T1 fat TTE with agitated
evaluation TEE TEE suppression saline
Consider diagnostic Renal artery
cerebral angiogram ultrasound if concern
to evaluate for for FMD
mycotic aneurysms
Notes See page 89 for For further
management management see
page 114
HACEK Haemophilus, Aggregatibacter actinomycetemcomitans, Cardiobacterium hominis, Eikenella
corrodens, Kingella kingae, TTE trans-thoracic echocardiography, TEE transesophageal echocardiog-
raphy, SLE systemic lupus erythematosus, FMD fibromuscular dysplasia, PFO patent foramen ovale,
ASD atrioseptal defect
82
Fig. 13.1 Paradoxical Embolus: Transesophageal echocardiogram showing a large clot traversing
a PFO This patient presented with an acute R MCA stroke in the context of prolonged immobility
after orthopedic surgery. Further workup demonstrated multiple DVT and transesophageal ECHO
demonstrated this finding
83
ISCHEMIA DUE TO VASCULOPATHY
VASCULITIS DUE TO INFECTIOUS
NONINFECTIOUS VASCULITIS [6] SYSTEMIC DISEASE VASCULITIS
Etiology Large vessel: Takayasu & Giant Cell SLE HIV (human
Arteritis (GCA) Rhumatoid arthritis immune-deficiency
Medium vessel: Polyarteritis nodosa & (RA) virus)
Kawasaki disease (present in children) Sjogrens VZV (Varicella
Small vessel: ANCA-associated, Solid organ Zoster Virus)
immune complex-mediated, neoplasms Bacterial meningitis
cryoglobulinemia Clonal B cell
lympho-proliferative
disorders
Behcet’s disease
Clues Dependent on syndrome, but Dependent on Dependent on
generally: constitutional symptoms, syndrome, often infection, assess for
athralgias, hypertension constitutional nuchal rigidity
For GCA: temporal pain, jaw symptoms
claudication, elderly patient, strokes
uncommon but predilection for
posterior circulation
Lab evaluation ESR/CRP, ANCA, cryoglobulins, ESR/CRP, ANA, Lumbar puncture (LP)
complement level dsDNA, RF, SS-A, for CSF cultures
SS-B, RNP, anti-smith Blood cultures
HIV antibody test
CSF VZV IgG/IgM
and PCR
Further CTA of great vessels Sjogrens: parotid
investigations Ultrasound and biopsy of temporal gland biopsy
artery if GCA suspected Neoplasms: workup
Evaluation for HCV (hep C virus) if per above
cryoglobulin being considered Behcets: pathergy test
Notes Consider MR-vessel wall imaging
84
ISCHEMIA DUE TO VASCULOPATHY
PRIMARY VASCULITIS INTRAVASCULAR
OF THE CNS LYMPHOMA GENETIC VASCULOPATHIES [9]
Etiology Primary CNS Lymphoma cells Moya-moya
vasculitis (PACNS) proliferate in the Cerebral autosomal dominant
Susac’s syndrome vessel wall of small arteriopathy with subcortical infarcts
(categorized more blood vessels and leukoencephalopathy (CARASIL)
specifically as a rare CARASIL (recessive)
form of COL41A mutation [3]
microangiopathy) Fabry’s disease
Hereditary Hemorrhagic
Telangiectasia (HHT)
Fibromuscular displasia (FMD)
Retinal vasculopathy with Cerebral
Leukodystrophy
Clues Primary CNS Subacute Moya-moya: progressive stenosis of
vasculitis often encephalopathy, the terminal and proximal ACA/
presents with subcortical infarcts, MCA. Vessels have a “puff of smoke
subacute constitutional B appearance” (see image below) [4]
encephalopathy + symptoms, skin CADASIL: family history of migraines,
headache lesions early onset strokes, dementia and
Susac’s syndrome is depression, subcortical infarcts
the triad of Fabry: Posterior circulation strokes,
encephalopathy, neuropathy, painful acroparasethesias
branched retinal CARASIL: subcortical infarcts
artery occlusion, and
hearing loss
Lab evaluation Rule out of other CSF for cytology and NOTCH3 (CADASIL)
causes of vasculitis flow cytometry and Fabry: Leukocyte alpha-galactosidase
IgH gene A activity
rearrangement
HTRA1 (CARASIL)
Retinal vasculopathy
Cerebral Leukodystrophy: TREX1
Further Brain biopsy for Dermatology consult MRI can demonstrate the pulvinar
investigations PACNS for skin biopsy sign in Fabry’s disease
Fluorescein PET scan to detect MRI shows temporal pole
angiogram for Susac subclinical systemic leukoaraiosis in CADASIL
lymphoma
Notes Consider MR-vessel wall imaging
ANCA antineutrophil cytoplasmic antibodies; ANA antinuclear antigen; dsDNA double stranded
DNA antibody; RF rheumatoid factor; SS-A Anti-Ro antibodies; SS-B Anti-La antibodies; RNP ribonu-
cleoprotein antibodies; PET positron emission tomography
85
Fig. 13.2 Moya-Moya: Patient demonstrates the characteristic loss of large vessels at the circle
of Willis (absence of MCA demonstrated by the arrow) and “puff of smoke” abnormal vasculature
(depicted by arrowheads)
86
OTHER ETIOLOGIES OF ISCHEMIA
REVERSIBLE CEREBRAL POSTERIOR REVERSIBLE
VASOCONSTRICTION ENCEPHALOPATHY SYNDROME
SYNDROME (RCVS) [5] (PRES) [5] ANEURYSMAL SAH
Etiology Unknown Failure of cerebral vaso- Can result in delayed
pathophysiology – autoregulation and endothelial cerebral ischemia
altered vascular tone and dysfunction
vasomotor control are
invoked
Clues Recurrent thunderclap Headache accompanied by Higher grades of SAH
headache encephalopathy +/− vision have increasing
Associated drugs: SSRIs, changes. incidence of delayed
cocaine, amphetamines, Accompanying conditions: cerebral ischemia and
diet pills hypertension, pregnancy clinically significant
Associated conditions: (hypertensive encephalopathy and vasospasm (see page
Pregnancy, migraine eclampsia are related conditions 235)
that may share the same
pathophysiology)
Precipitating medications:
immunomodulators (tacrolimus,
sirolimus), chemotherapeutic
agents
Lab Urine and serum drug Urine and serum drug screen, drug N/A
evaluation screen levels may be helpful in
appropriate cases
Imaging CTA demonstrating MRI demonstrating posterior TCD, cvEEG, CTA, and
evaluation vasospasm dominate leukoencephalopathy angiography as
warranted
Notes More commonly causes Strokes are an infrequent See page 229 for
convexity SAH than complication, posterior cerebral prevention and
ischemic infarcts edema without infarct is typical management
SSRIs selective serotonin reuptake inhibitor, SAH subarachnoid hemorrhage, TCDs transcranial doppler,
cvEEG continue video electroencephalogram
REFERENCES
1. Jeon S-B, et al. Homocysteine, small-vessel disease, and atherosclerosis: an MRI study of 825
stroke patients. Neurology. 2014;83(8):695–701.
2. Moore GW, et al. Further evidence of false negative screening for lupus anticoagulants. Thromb
Res. 2008;121(4):477–84.
3. Alamowitch S, et al. Cerebrovascular disease related to COL4A1 mutations in HANAC syn-
drome. Neurology. 2009;73(22):1873–82.
87
4. Fukui M, et al. Moyamoya disease. Neuropathology. 2000;20:61–4.
5. Pilato F, Distefano M, Calandrelli R. Posterior reversible encephalopathy syndrome and revers-
ible cerebral vasoconstriction syndrome: clinical and radiological considerations. Front Neurol.
2020;11:34.
6. Moore PM. Vasculitis of the central nervous system. Curr Rheumatol Rep. 2000;2(5):376–82.
7. Voetsch B, et al. Inherited thrombophilia as a risk factor for the development of ischemic stroke
in young adults. Thromb Haemost. 2000;83(02):229–33.
8. Ji R, et al. Ischemic stroke and transient ischemic attack in young adults: risk factors, diagnostic
yield, neuroimaging, and thrombolysis. JAMA Neurol. 2013;70(1):51–7.
9. Putaala J. Ischemic stroke in young adults. Continuum (Minneap Minn). 2020;26(2):386–414.
https://doi.org/10.1212/CON.0000000000000833.
88
ISCHEMIC STROKE: DISSECTION
Catherine S. W. Albin and Sahar F. Zafar
Both
□□History of trauma
□□History of connective tissue disease: fibromuscular dysplasia, Ehlers Danlos
type IV, Marfan’s, cystic medial necrosis, osteogenesis imperfecta, polycystic
kidney disease
□□ Systemic cause of vessel wall inflammation: infectious or rheumatologic/
autoimmune conditions
□□ Stroke in the absence of traditional vascular risk factors
□□ Young age (patients average ~40 years old in case series)
Internal Carotid
□□Headache/facial pain – may be referred to supraorbital ridge
□□Neck pain – commonly along the anterolateral neck
□□Partial Horner’s syndrome – ipsilateral miosis and ptosis
□□Anterior circulation stroke syndrome
Vertebral Artery
□□Headache – often ipsilateral occipital
□□Neck pain – commonly posterior lateral neck
□□Cerebellar or brainstem symptoms
“Flame sign”
Pathophysiology of dissection
(tapering of the ICA)
ADMISSION CHECKLIST
□□ Review vessel imaging
• Angiography is the gold standard, but infrequently performed given procedural
risks; CT angiography is preferable when GFR >35.
• MRA TOF may overcall the degree of vessel narrowing and occlusion as this
is a flow-dependent study.
• MRI T1 Fat Suppression can help to highlight the vessel wall hematoma.
Being stationary blood, the hematoma will be T1 hyperintense.
□□ Determine if the dissection is extra- or intracranial as this may impact manage-
ment decisions (see below)
• Traditional teaching was that given the risk of subarachnoid hemorrhage with
sub-adventitial extension, dissections that were even partially intracranial
should not be treated with anticoagulation.
• However, a case-series of 81 patients with intracranial dissections (carotid or
vertebral) with no SAH at the time of presentation, who were subsequently
treated with anticoagulation, found no events of SAH during treatment [2].
□□ Review parenchymal imaging
• DWI MRI preferable to determine the size of ischemic core
• SWI MRI may be helpful to exonerate SAH
□□ Provide adequate analgesia
□□ Determine use of aspirin vs anticoagulation (see below)
90
DETERMINING WHEN A VESSEL IS EXTRACRANIAL VS. INTRACRANIAL
Carotids
• The carotid artery is named based on the segment classification proposed by
Bouthillier [3].
• It is considered intracranial when it enters the skull base at the petrous portion.
• The carotid is not intradural until passing the distal dural ring, which anatomically
demarcates the end of the clinoid (C5) portion.
• Distal to the dural ring, there is a risk of SAH with dissection extension. However,
subadventitial dissection in the cavernous segment (C4) could result in a
cavernous-carotid fistula, which would also have serious consequences.
• While intradural dissections do result in SAH, the likelihood that subarachnoid
hemorrhage develops as a result of treatment is small and has not been well-
documented in case-series [2].
Vertebrals
• Pierces the dura after passing through the transverse foramen and the posterior
atlanto-occipital membrane of C1.
C7 – Communicating
Distal Dural C6 – Ophthalmic
Ring V4 –
C5 – Clinoid Intradural
C4 - Cavernous V3 –
C2 to Dura
C3 - Lacerum
C2 - Petrous V2 - Foraminal
C1 – Cervical
V1 – pre-
foraminal
91
ASPIRIN VS. ANTICOAGULATION
TRIAL METHODS FINDINGS
CADISS [4] 250 patients with extracranial Anti-platelet therapy was
(Lancet 2014) vertebral or carotid dissections non-inferior, but the rate of stroke
randomized within the first 7 days was only 2%, which was
of symptom discovery to anti- significantly lower in both groups
platelet treatment (aspirin, than expected.
ASA + clopidogrel, dipyridamole
at the discretion of the physician)
or anticoagulation.
Take away from CADISS: The trial was underpowered and the antiplatelet group was not
standardized. Additionally, patients were randomized at a mean of 3.25 days after symptom onset,
which misses the highest risk period for stroke when anticoagulation might prove more efficacious.
There is still considerable practice variation, anticoagulation (most often as heparin to warfarin) or
antiplatelet (most often ASA 325 mg) can be justified on a case-by-case basis.
TREAT-CAD [5] 194 patients with cervical artery Non-inferiority of aspirin was not
(Lancet 2021) dissection (not specified intra- or found, suggesting superiority of
extracranial) within 2 weeks anticoagulation although the trial
before enrollment were was NOT designed to prove
randomized to 300 mg of aspirin that. The observed absolute
or a vitamin K antagonist for difference in the primary
90 days. Used composite primary endpoint rate between groups
endpoints: clinical events (stroke, was 8%. Ischemic stroke, major
major hemorrhage, death) and hemorrhage, or MRI surrogates
MRI outcomes at 14 days. occurred in 23% in the ASA
group and 15% in the Vitamin K
group.
Take away from TREAT-CAD: This trial was not designed to show the superiority of a vitamin K
antagonist, but it was not able to demonstrate non-inferiority of aspirin. Unfortunately, two commonly
used practices: dual-antiplatelet therapy and direct oral anticoagulants were not investigated. Some
trial notes for consideration: 13% of patients were treated with acute revascularization prior to
enrollment. The time between onset of first dissection symptom and treatment was, on average,
7 days in both groups.
92
REFERENCES
1. Silbert PL, Mokri B, Schievink WI. Headache and neck pain in spontaneous internal carotid and
vertebral artery dissections. Neurology. 1995;45(8):1517–22.
2. Metso TM, Metso AJ, Helenius J, Haapaniemi E, Salonen O, Porras M, Hernesniemi J, Kaste M,
Tatlisumak T. Prognosis and safety of anticoagulation in intracranial artery dissections in adults.
Stroke. 2007;38(6):1837–42.
3. Bouthillier A, van Loveren HR, Keller JT. Segments of the internal carotid artery: a new classifica-
tion. Neurosurgery. 1996;38(3):425–32; discussion 432–3.
4. CADISS Trial Investigators. Antiplatelet treatment compared with anticoagulation treatment for
cervical artery dissection (CADISS): a randomised trial. Lancet Neurol. 2015;14(4):361–7.
5. Engelter ST, et al. TREAT-CAD Investigators. Aspirin versus anticoagulation in cervical artery
dissection (TREAT-CAD): an open-label, randomised, non-inferiority trial. Lancet Neurol.
2021;20(5):341–50. https://doi.org/10.1016/S1474-4422(21)00044-2. Epub ahead of print.
93
ISCHEMIC STROKE: SYMPTOMATIC CAROTID STENOSIS
(“HOT CAROTID”)
Catherine S. W. Albin and Sahar F. Zafar
“SYMPTOMATIC” DEFINITION
A focal, acute event (stroke, TIA, or amaurosis fugax) resulting from a perfusion
deficit or embolic event in the territory of an internal carotid artery with significant
atherosclerotic burden (at least 50% stenosis by NASCET criteria). Note that patients
with moderate or severe strokes (with persistent disabling neurologic deficits) have
been excluded from trials of carotid artery stenting (CAS)/carotid endarterectomy
(CEA). For many trials, patients were considered symptomatic if an index event had
happened within 180 days prior to treatment.
96
TIMING OF SURGERY
The risk of neurologic worsening is estimated between 8% and 27% in the first
2 weeks [10]. Multiple studies have confirmed that the benefit of CEA is most appar-
ent if offered within the first 2 weeks of index symptom. The periprocedural stroke
risk of CAS is higher within the first 2 weeks after index symptom, up to 9.4% [11].
Although management is often institution and operator-dependent, CEA may be
preferable in the urgent setting.
97
REFERENCES
1. Moneta GL, et al. Correlation of North American Symptomatic Carotid Endarterectomy Trial
(NASCET) angiographic definition of 70% to 99% internal carotid artery stenosis with duplex
scanning. J Vasc Surg. 1993;17(1):152–9.
2. Howard VJ, et al. Carotid revascularization and medical management for asymptomatic carotid
stenosis: protocol of the CREST-2 clinical trials. Int J Stroke. 2017;12(7):770–8.
3. Sardar P, Chatterjee S, Aronow HD, Kundu A, Ramchand P, Mukherjee D, et al. Carotid artery
stenting versus endarterectomy for stroke prevention: a meta-analysis of clinical trials. J Am Coll
Cardiol. 2017;69(18):2266–75.
4. Batchelder AJ, Hunter J, Robertson V, et al. Dual antiplatelet therapy prior to expedited carotid
surgery reduces recurrent events prior to surgery without increasing peri-operative bleeding com-
plications. Eur J Vasc Endovasc Surg. 2015;50:412–9.
5. Amarenco P, et al. Benefit of targeting a LDL (low-density lipoprotein) cholesterol <70 mg/dL dur-
ing 5 years after ischemic stroke. Stroke. 2020;51(4):1231–9.
6. Martinez-Gutierrez, Carlos J, et al. Preoperative antithrombotic treatment in acutely symptomatic
carotid artery stenosis. Journal of Stroke and Cerebrovascular Diseases 31.5 (2022): 106396.
7. Salem MK, et al. Spontaneous cerebral embolisation in asymptomatic and recently symptomatic
patients with TIA/Minor stroke. Eur J Vasc Endovasc Surg. 2011;41(6):720–5.
8. Altaf N, et al. Detection of intraplaque hemorrhage by magnetic resonance imaging in symptom-
atic patients with mild to moderate carotid stenosis predicts recurrent neurological events. J Vasc
Surg. 2008;47(2):337–42.
9. Prabhakaran S, et al. Carotid plaque surface irregularity predicts ischemic stroke: the northern
Manhattan study. Stroke. 2006;37(11):2696–701.
10. Ois A, Cuadrado-Godia E, Rodríguez-Campello A, Jimenez-Conde J, Roquer J. High risk of early
neurological recurrence in symptomatic carotid stenosis. Stroke. 2009;40(8):2727–31.
11. Rantner B, Goebel G, Bonati LH, Ringleb PA, Mas JL, Fraedrich G, Carotid Stenting Trialists’
Collaboration. The risk of carotid artery stenting compared with carotid endarterectomy is great-
est in patients treated within 7 days of symptoms. J Vasc Surg. 2013;57(3):619–26.
98
ISCHEMIC STROKE – POST STROKE MANAGEMENT
OF ANTICOAGULATION
Catherine S. W. Albin and Sahar F. Zafar
14 12.2
12 11.2 10.8
9.7
10
8 7.2
6 4.8
4 3.2
2.2
2 0 0.6
0
0 1 2 3 4 5 6 7 8 9
CHA2DS2-Vasc Score
Chart above demonstrating the annualized stroke risk, unadjusted for aspirin use
based on the Swedish Atrial Fibrillation cohort study [3].
Mechanical Valves
In a large meta-analysis of complications of mechanical valves, the annualized risk of
a “Major Embolism” (classified as one causing death, persistent neurologic deficit or
peripheral disease requiring surgery) was 4 per 100 patient-years among all patients
with any mechanical value when not on antithrombotic therapy [5]. The RR of
embolism was twice as high with a valve in the mitral position. With coumadin the
risk dropped to 1.4 per 100 patient years.
1
Adams Jr, Harold P., et al. “Classification of subtype of acute ischemic stroke. Definitions for use in
a multicenter clinical trial. TOAST. Trial of Org 10172 in Acute Stroke Treatment.” stroke 24.1
(1993): 35-41.
100
PART 2: UNDERSTANDING THE PATIENT’S PERSONAL RISK OF BLEEDING
The HeRS score for predicting hemorrhagic transformation is based on this data
and can be helpful in determining risk
(Available as an iPhone app “Johns Hopkins HeRS”)
101
Grading Hemorrhagic Conversion
Proposed for use in the ECASS trial [10]
HEMORRHAGIC
CLASSIFICATION RADIOGRAPHIC APPEARANCE EXAMPLE
Hemorrhagic Small, hyperdense petechiae without
infarction type 1 mass effect
(HI1)
102
HEMORRHAGIC
CLASSIFICATION RADIOGRAPHIC APPEARANCE EXAMPLE
Parenchymal Homogeneous hyperdensity occupying
hematoma type 2 >30% of the infarct zone; significant
(PH2) mass effect; or, any homogenous
hyperdensity located beyond the
borders of the infarct zone
103
REFERENCES
1. Hart RG, Benavente O, McBride R, Pearce LA. Antithrombotic therapy to prevent stroke in
patients with atrial fibrillation: a meta-analysis. Ann Intern Med. 1999;131:492–501.
2. Lip GYH, et al. Refining clinical risk stratification for predicting stroke and thromboembolism in
atrial fibrillation using a novel risk factor-based approach: the euro heart survey on atrial fibrilla-
tion. Chest. 2010;137(2):263–72.
3. Friberg L, Rosenqvist M, Lip GYH. Evaluation of risk stratification schemes for ischaemic stroke
and bleeding in 182 678 patients with atrial fibrillation: the Swedish Atrial Fibrillation cohort study.
Eur Heart J. 2012;33(12):1500–10.
4. Saxena R, Lewis S, Berge E, Sandercock PAG, Koudstaal PJ, for the International Stroke Trial
Collaborative Group. Risk of early death and recurrent stroke and effect of heparin in 3169
patients with acute ischemic stroke and atrial fibrillation in the International Stroke Trial. Stroke.
2001;32:2333–7.
5. Cannegieter SC, Rosendaal FR, Brite E. Thromboembolic and bleeding complications in patients
with mechanical heart valve prostheses. Circulation. 1994;89:635–41.
6. Johansson E, et al. Recurrent stroke in symptomatic carotid stenosis awaiting revascularization:
a pooled analysis. Neurology. 2016;86(6):498–504.
7. Stromberg S, et al. Risk of early recurrent stroke in symptomatic carotid stenosis. Eur J Vasc
Endovasc Surg. 2015;49(2):137–44.
8. Marsh EB, Llinas RH, Hillis AE, et al. Hemorrhagic transformation in patients with acute ischemic
stroke and an indication for anticoagulation. Eur J Neurol. 2013;20:962–7.
9. Lee SH, et al. Predictors of hemorrhagic transformation in patients with mild atrial fibrillation-
associated stroke treated with early anticoagulation: post hoc analysis of the Triple AXEL Trial.
Clin Neurol Neurosurg. 2018;174:156–62.
10. del Zoppo GJ, Poeck K, Pessin MS, Wolpert SM, Furlan AJ, Ferbert A, Alberts MJ, Zivin JA,
Wechsler L, Busse O, Greenlee R Jr, Brass L, Mohr JP, Feldmann E, Hacke W, Kase CS, Biller
J, Gress D, Otis SM. Recombinant tissue plasminogen activator in acute thrombotic and embolic
stroke. Ann Neurol. 1992;32:78–86.
11. Paciaroni M, Giancarlo A, Micheli S, Caso V. Efficacy and safety of anticoagulation treat-
ment in acute cardioembolic stroke: a meta-analysis of randomized controlled trials. Stroke.
2007;38:423–30.
12. Hong KS, Kwon S, Lee SH. Rivaroxaban vs warfarin sodium in the ultra-early period after
atrial fibrillation-related mild ischemic stroke: a randomized clinical trial. JAMA Neurol.
2017;74(10):1206–15.
13. Heidbuchel, Hein, et al. EHRA practical guide on the use of new oral anticoagulants in patients
with non-valvular atrial fibrillation: executive summary. European heart journal 34.27 (2013):
2094–106.
104
SELECTED ANTI-PLATELETS AND ANTICOAGULATION
IN STROKE PREVENTION
Catherine S. W. Albin and Megan E. Barra
MECHANISM
NAME (ABBREVIATED) DOSING NOTES/TRIAL DATA
Anti-platelet drugs
Aspirin (ASA) Irreversible inhibition 325 mg or 81 mg PO QD CAST (Lancet 1997) [1]
of formation of IST (Lancet 1997) [2]
thromboxane A2;
↓platelet (plt)
aggregation
Clopidogrel Irreversible P2Y12 Variable in trials some load POINT (NEJM 2018) [3]
(Plavix) adenosine 300 or 600 mg PO ×1; CHANCE (NEJM 2013) [4]
diphosphate (ADP) followed by 75 PO QD SAMMPRIS (NEJM 2011) [5]
antagonists; ↓plt
aggregation
Ticagralor Reversibly binds ADP 180 mg PO ×1 followed by THALES (NEJM 2020) [6]
(Brilinta) P2Y12 receptor; ↓plt 90 mg PO BID
aggregation
Dipyridamole Dipyridamole is 200 mg PO BID ESPRIT (Lancet 2006) [7]. Use
+ ASA XR phosphodiesterase often limited by headache.
(Aggrenox) inhibitor
Anticoagulants
Warfarin Vitamin K antagonist Target INR usually 2–3 SPAF III (Circulation 1991)
(Coumadin) (decreased synthesis [13]
of factor II, VII, IX, X, ACTIVE W (Lancet 2006) [8]
and protein C and S)
Unfractionated Primarily potentiates High-intensity therapy often IST (Lancet 1997) [2]; Can
heparin action of ATIII used for thromboembolic monitor with anti-Xa if
inhibition of clotting disease. However, in the setting prolonged baseline
factors (primarilyXa/ of acute stroke a low-intensity PTT (e.g. lupus anticoagulant)
IIa) goal may be targeted. or pseudo-heparin resistance
Prevents fibrin (elevated VIII or fibrinogen)
Post-stroke often dosed
formation
without boluses. Follow Short duration of action:
institution-specific dosing and T1/2 = 60–90 min
titration protocols for PTT
and/or anti-Xa targets
T1/2: 5–13 h
106
MECHANISM
NAME (ABBREVIATED) DOSING NOTES/TRIAL DATA
Apixaban Factor Xa inhibitor 5 mg PO BID Not well studied in CKD
(Eliquis)
Dose reduce if ≥ 2 patient ROCKET-AF (NEJM 2011)a
criteria met: ≥80 yo, ≤60 kg, [12]
Cr ≥ 1.5, reduce dose to
T1/2: 8–15 h
2.5 mg BID
a
Trial compared the oral anticoagulant to warfarin to demonstrate non-inferiority in stroke pre-
vention for non-valvular atrial fibrillation
REFERENCES
1. Chen Z-M. CAST: randomised placebo-controlled trial of early aspirin use in 20 000 patients with
acute ischaemic stroke. Lancet. 1997;349(9066):1641–9.
2. International Stroke Trial Collaborative Group. The International Stroke Trial (IST): a randomised
trial of aspirin, subcutaneous heparin, both, or neither among 19 435 patients with acute isch-
aemic stroke. Lancet. 1997;349(9065):1569–81.
3. Johnston SC, et al. Clopidogrel and aspirin in acute ischemic stroke and high-risk TIA. N Engl J
Med. 2018;379(3):215–25.
4. Wang Y, et al. Clopidogrel with aspirin in acute minor stroke or transient ischemic attack. N Engl
J Med. 2013;369(1):11–9.
5. Chimowitz MI, et al. Stenting versus aggressive medical therapy for intracranial arterial stenosis.
N Engl J Med. 2015;365(11):993–1003.
6. Johnston SC, et al. Ticagrelor and aspirin or aspirin alone in acute ischemic stroke or TIA. N Engl
J Med. 2020;383:207–17.
7. Halkes PH, et al. Aspirin plus dipyridamole versus aspirin alone after cerebral ischaemia of arte-
rial origin (ESPRIT): randomized controlled trial. Lancet. 2006;367(9523):1665–73.
8. Connolly SJ, et al. Clopidogrel plus aspirin versus oral anticoagulation for atrial fibrillation in the
Atrial fibrillation Clopidogrel Trial with Irbesartan for prevention of Vascular Events (ACTIVE W):
a randomised controlled trial. Lancet. 2006;367(9526):1903–12.
9. Navi BB, et al. Enoxaparin vs aspirin in patients with cancer and ischemic stroke: the TEACH pilot
randomized clinical trial. JAMA Neurol. 2018;75(3):379–81.
10. Connolly SJ, et al. Dabigatran versus warfarin in patients with atrial fibrillation. N Engl J Med.
2009;361(12):1139–51.
11. Granger CB, et al. Apixaban versus warfarin in patients with atrial fibrillation. N Engl J Med.
2011;365(11):981–2.
12. Patel MR, et al. Rivaroxaban versus warfarin in nonvalvular atrial fibrillation. N Engl J Med.
2011;365(10):883–91.
13. SPAF Investigators. Stroke prevention in atrial fibrillation study. Final results. Circulation.
1991;84(2):527–39.
107
ACUTE MANAGEMENT STRATEGIES: tPA AND MECHANICAL
THROMBECTOMY TRIALS
Catherine S. W. Albin and Sahar F. Zafar
110
TRIAL TRIAL DESIGN MAJOR FINDINGS
DEFUSE 3 Patients with ICA/prox M1 At 90 days, 45% of patients in the
(NEJM 2018) [10] occlusion presenting 6–16 h clot retrieval group were functionally
from LSW with infarct <70 mL independent (mRS 0–2) vs. just 17%
and perfusion imaging showing in the medical management
an ischemic tissue to infarcted
No difference in ICH
tissue ratio of 1.8 were
randomized to clot retrieval vs.
medical management
Mechanical thrombectomy has revolutionized stroke care for patients with large vessel occlusion.
Pooled data from DAWN and DEFUSE-3 has reinforced that endovascular therapy is superior to
medical management in patients with AIS from LVO beyond 6 h of LSW, if appropriately selected
[11]
Selected tPA or no-tPA prior to mechanical thrombectomy (MT) – (“bridging” trials)
DIRECT MT Patients either treated with MT Non-inferiority (changed from
(NEJM 2020) [12] alone (n = 326) or IV tPA superiority). Non-inferior for a mRS @
(<4.5 h, standard dose) 90 days (20% margin). Lower
(n = 328). Included patients with successful reperfusion before
ICA, M1, M2 occlusions thrombectomy (2.4% vs. 7%) and
lower successful reperfusion (79.4%
vs. 84.5%) when no IV tPA given. No
stat sig change in ICH or 90-day
mortality with or without tPA
NIHSS ≥ 6
symptomatic ICH was not statistically
different
111
TRIAL TRIAL DESIGN MAJOR FINDINGS
DEVT Trial Patients either treated with MT Non-inferiority trial set at 10% for
(JAMA 2021) [14] alone (n = 116) or IV tPA+ MT achieving 90-day functional
(n = 118), standard tPA dose ≤
independence (mRA 2). Trial
given, and the full infusion was stopped early as the interim analysis
given even if reperfusion (20% of total sample, 194 patients)
happened during the infusion demonstrated non-inferiority
112
Meta-analysis of randomized controlled trials [18] comparing the outcomes of early initiation of
short-term DAPT (aspirin + a P2Y12 inhibitor for up to 3 months) vs. aspirin monotherapy
demonstrated that patients treated with DAPT had a lower risk of recurrent stroke (RR 0.76 [95% CI
0.68–0.83]; P < 0.01), but a higher risk of major bleeding events (RR 2.22 [95% 1.14–4.34])
EXTRACRANIAL DISEASE
NASCET Patients age <80 w/ 26% of those in the medical treatment
(NEJM 1991) [19] extracranial ICA stenosis group had a recurrent stroke in 2
70–99% were randomized to years vs. 9% in CEA group.
medical therapy (ASA 1300 mg Preoperative morbidity ~6%. NASCET
+ lipid/dm/HTN control) vs. II demonstrated some efficacy of CEA
medical therapy + Carotid in patients w/ ICA stenosis 50–69%.
Endarterectomy (CEA) Note that statsin were not in wide use
at the time and medical tx was not
standardized
CREST Patients with symptomatic or There was no difference in composite
(NEJM 2010) [20] asymptomatic carotid stenosis outcomes (stroke, death, MI) between
(variably measured, but >70% the two treatment arms; however,
on carotid U/S for both groups) there was a higher rate of
randomized to CEA or stenting. periprocedural stroke in the stenting
DAPT given prior to procedure group, and higher rate of MI in the
CEA group
CREST II Patients with asymptomatic Unknown as still enrolling. Important
(Enrolling) carotid stenosis (>70% on U/S) to know as this trial as asymptomatic
randomized to medical carotid disease can be treated with
management + CEA or CAS vs. interventional treatments due to
medical management alone clinical equipoise at this point
LDL Targets after Patients with ischemic stroke in 2860 patients were enrolled and
Ischemic Stroke previous 3 months or TIA in followed for median 3.5 years.
(NEJM 2020) [21] preceding 15 days assigned to Stopped early. 8.5% endpoints in
LDL target of <70 mg/dL or <70 mg/dL group and 10.9% in the
target 90–110 mg/dL. All higher target group. 34% of patients
patients had evidence of in the <70 mg/dL group required
cerebrovascular or coronary ezetimibe in addition to statins
artery diease (CAD). Outcome No statistically significant difference
was major cardiovascular event between new diabetes or ICH
(stroke, MI, coronary or carotid although a numerically higher number
revascularization) or death of ICH in the lower-target group
113
SEVERE INTRACRANIAL STENOSIS
WASID Patients with recent TIA or Stopped early because increased
(NEJM 2005) [22] non-disabling stroke with hemorrhage in the warfarin group.
intracranial stenosis of >50% Subgroup analysis demonstrated
randomized to either warfarin or lower primary outcome (not just
1300 mg ASA strokes) in the basilar subgroup; those
with therapeutic INRs in the basilar
group may have had lower stroke risk
SAMMPRIS Patients with recent TIA or Medical tx: Aspirin 325 mg + Plavix
(NEJM 2011) [23] non-disabling stroke with 75 mg for 90 days + “lifestyle
intracranial stenosis of >70% modification program” resulted in
randomized to either medical tx significantly less strokes than stenting
or medical tx + stenting at 1 year
114
RESPECT 980 patients, ≤60 years old w/ The PFO closure group was
(NEJM 2017) [26] cryptogenic stroke within 270 associated with 0.58 ischemic events
days prior to randomization and per 100 patient-years vs. 1.07 per
PFO confirmed by TTE. Patients 100 patient- years in the medical
randomized 1:1 PFO + management group. (P = 0.046).
antiplatelet vs. medical Estimated NNT of 42 to prevent 1
management (ASA, warfarin, stroke within 5 years
clopidogrel, ASA+dipyridamole)
A meta-analysis of randomized controlled trials [27] comparing PFO closure with medical therapy
(anticoagulation or antiplatelet therapy) for cryptogenic stroke demonstrated superiority of PFO
closure to prevent stroke recurrence; however, the annual absolute risk reduction was low. PFO
closure was associated with an increased risk of atrial fibrillation
REFERENCES
1. The National Institute of Neurological Disorders and Stroke rt-PA Stroke Study Group. Tissue
plasminogen activator for acute ischemic stroke. N Engl J Med. 1995;333:1581–7.
2. Bluhmki E, et al. Stroke treatment with alteplase given 3· 0–4· 5 h after onset of acute ischaemic
stroke (ECASS III): additional outcomes and subgroup analysis of a randomised controlled trial.
Lancet Neurol. 2009;8(12):1095–102.
3. Thomalla G, et al. MRI-guided thrombolysis for stroke with unknown time of onset. N Engl J Med.
2018;379(7):611–22.
4. Furlan A, et al. Intra-arterial prourokinase for acute ischemic stroke: the PROACT II study: a ran-
domized controlled trial. JAMA. 1999;282(21):2003–11.
5. Broderick JP, Palesch YY, Demchuk AM, et al. Endovascular therapy after intravenous t-PA ver-
sus t-PA alone for stroke. N Engl J Med. 2013;368:893–903.
6. Berkhemer OA, Fransen PS, Beumer D, et al. A randomized trial of intraarterial treatment for
acute ischemic stroke. N Engl J Med. 2015;372:11–20.
7. Goyal M, et al. Randomized assessment of rapid endovascular treatment of ischemic stroke. N
Engl J Med. 2015;372(11):1019–30.
8. Campbell BC, Mitchell PJ, Kleinig TJ, et al. Endovascular therapy for ischemic stroke with
perfusion-imaging selection. N Engl J Med. 2015;372:1009–18.
9. Nogueira RG, Jadhav AP, Haussen DC, et al. Thrombectomy 6 to 24 hours after stroke with a
mismatch between deficit and infarct. N Engl J Med. 2018;378:11–21.
10. Albers GW, et al. Thrombectomy for stroke at 6 to 16 hours with selection by perfusion imaging.
N Engl J Med. 2018;378(8):708–18.
11. Snelling B, et al. Extended window for stroke thrombectomy. J Neurosci Rural Pract.
2019;10(2):294–300. https://doi.org/10.4103/jnrp.jnrp_365_18.
12. Yang P, Zhang Y, Zhang L, Zhang Y, Treurniet KM, Chen W, Peng Y, Han H, Wang J, Wang
S, DIRECT-MT Investigators, et al. Endovascular thrombectomy with or without intravenous
alteplase in acute stroke. N Engl J Med. 2020;382:1981–93.
13. Suzuki K, et al. The randomized study of endovascular therapy with versus without intravenous
tissue plasminogen activator in acute stroke with ICA and M1 occlusion (SKIP study). Int J Stroke.
2019;14(7):752–5.
115
14. Zi W, et al. Effect of endovascular treatment alone vs intravenous alteplase plus endovascular
treatment on functional independence in patients with acute ischemic stroke: the DEVT random-
ized clinical trial. JAMA. 2021;325(3):234–43.
15. Wang Y, et al. Clopidogrel with aspirin in acute minor stroke or transient ischemic attack. N Engl
J Med. 2013;369:11–9.
16. Johnston SC, et al. Clopidogrel and aspirin in acute ischemic stroke and high-risk TIA. N Engl J
Med. 2018;379(3):215–25.
17. Johnston SC, et al. Ticagrelor and aspirin or aspirin alone in acute ischemic stroke or TIA. N Engl
J Med. 2020;383(3):207–17.
18. Bhatia K, et al. Dual antiplatelet therapy versus aspirin in patients with stroke or transient isch-
emic attack: meta-analysis of randomized controlled trials. Stroke. 2021;52(6):e217–23.
19. North American Symptomatic Carotid Endarterectomy Trial Collaborators*. Beneficial effect of
carotid endarterectomy in symptomatic patients with high-grade carotid stenosis. N Engl J Med.
1991;325(7):445–53.
20. Brott TG, et al. Stenting versus endarterectomy for treatment of carotid-artery stenosis. N Engl J
Med. 2010;363(1):11–23.
21. Amarenco P, et al. A comparison of two LDL cholesterol targets after ischemic stroke. N Engl J
Med. 2020;382(1):9–19.
22. Chimowitz MI, et al. Comparison of warfarin and aspirin for symptomatic intracranial arterial ste-
nosis. N Engl J Med. 2005;352(13):1305–16.
23. Derdeyn CP, et al. Aggressive medical treatment with or without stenting in high-risk patients
with intracranial artery stenosis (SAMMPRIS): the final results of a randomised trial. Lancet.
2014;383(9914):333–41.
24. Søndergaard L, et al. Patent foramen ovale closure or antiplatelet therapy for cryptogenic stroke.
N Engl J Med. 2017;377(11):1033–42.
25. Mas J-L, et al. Patent foramen ovale closure or anticoagulation vs. antiplatelets after stroke. N
Engl J Med. 2017;377(11):1011–21.
26. Saver JL, et al. Long-term outcomes of patent foramen ovale closure or medical therapy after
stroke. N Engl J Med. 2017;377(11):1022–32.
27. Turc G, et al. Closure, anticoagulation, or antiplatelet therapy for cryptogenic stroke with pat-
ent foramen ovale: systematic review of randomized trials, sequential meta-analysis, and new
insights from the CLOSE study. J Am Heart Assoc. 2018;7(12):e008356. https://doi.org/10.1161/
JAHA.117.008356.
116
VENOUS SINUS THROMBOSIS
Catherine S. W. Albin and Sahar F. Zafar
Cortical veins
Inferior sagittal
Transverse
sinus
Internal julgar
□□Female gender
□□Risk factors for hypercoagulable state [1]:
□□Personal or family history of blood clots
□□Pregnancy, post-partum, oral contraceptives (particularly high estrogen)
□□Malignancy
□□Systemic inflammatory diseases: particularly SLE, APLS, nephrotic syn-
drome, inflammatory bowel disease, and hematologic diseases (DIC, HIT,
JAK2 mutations, etc.)
□□ Dehydration
□□ Trauma to or neurosurgical manipulation near the sinuses
□□ Sinus infection (particularly if concern for a cavernous sinus thrombosis)
Note that in ISCVT, 44% of the patients had more than one cause or predisposing
factor. Genetic thrombophilias were present in 22% of patients [2].
Symptoms
□□Headache
□□Altered mental status
□□Cranial neuropathy of II, III, IV, VI (nerves that run in the cavernous sinus)
□□Seizure/post-ictal state (in a large series about 1/3 of patients had early
seizures) [3]
□□Focal neurologic symptoms if stroke, hemorrhage or edema develop
Neuroimaging Findings That Should Raise Concern for VST
Note: Non-contrast head CT may be normal in nearly half of the cases [4]
□□Cortical infarcts, particularly those with hemorrhagic transformation
□□Deep infarcts
□□Bilateral infarcts
□□Cortical SAH
□□CNS tumor near the sinuses
□□See examples on page 120
118
WORKUP AND TREATMENT
Workup [5]
□□CT Venography (less sensitive for thrombosis in smaller veins and corti-
cal veins)
□□ MRI and MR Venography (more sensitive for detecting thrombosis and impact
on the surrounding parenchyma)
□□ Workup for thrombophilia should be considered in collaboration with
hematology.1
□□ Search for underlying malignancy, infection, or inflammatory condition as
indicated by clinical circumstances
Acute Treatment
□□Anticoagulation: often IV unfractionated heparin due to the ability to titrate the
dose, target a specific level using either PTT or anti-Xa levels, and reverse if
needed. Studies suggest that acute anticoagulation does not seem to worsen
VST-related ICH [6].
□□ Frequent neuroimaging as therapeutic targets are met or for any new symptoms
□□ Management of elevated ICP (see page 187)
□□ Seizure treatment as indicated [7]
□□ Hydration
□□ Endovascular treatment for target clot lysis or extraction in very limited
circumstances.
Chronic Treatment
Duration of treatment is determined by the likelihood of re-thrombosis and underlying
risk factors. Vitamin K antagonism was the mainstay of treatment prior to direct oral
anticoagulants (DOACs). RE-SPECT CVT [8] was the first open-label randomized
trial to compare rates or bleeding and recurrent CVT and VTE in 120 VST patients
treated with warfarin versus dabigatran.
Over the 25 months of observation there was no recurrence of VTE in either group,
no worsening of CVT in either group. Bleeding was a complication in 20% of patients
in both groups. Of the patients that had baseline ICH, one new major bleed occurred
in the warfarin group and one patient in the dabigatran group had significant worsen-
ing of bleed.
This was a small trial, but it did suggest non-inferiority of a DOAC to warfarin.
Importantly, though, warfarin is still preferred for clots associated with antiphospho-
lipid antibodies.
1
Given that acute thrombosis disrupts the coagulation cascade, many tests that eval for underlying
thrombophilia are inaccurate in the acute period. Deferred testing may be recommended.
119
MRV demonstrating loss of opacification along CTV demonstrating filling defect within the
the superior sagittal sinus superior sagittal sinus
120
REFERENCES
1. Dentali F. Thrombophilic abnormalities, oral contraceptives, and risk of cerebral vein thrombosis:
a meta-analysis. Blood. 2006;107(7):2766–73.
2. Ferro JM, et al. Prognosis of cerebral vein and dural sinus thrombosis: results of the International
Study on Cerebral Vein and Dural Sinus Thrombosis (ISCVT). Stroke. 2004;35(3):664–70.
3. Ferro JM, et al. Seizures in cerebral vein and dural sinus thrombosis. Cerebrovasc Dis.
2003;15(1–2):78–83.
4. Kumral E, Polat F, Uzunköprü C, Çallı C, Kitiş Ö. The clinical spectrum of intracerebral hema-
toma, hemorrhagic infarct, non-hemorrhagic infarct, and non-lesional venous stroke in patients
with cerebral sinus–venous thrombosis. Eur J Neurol. 2012;19:537–43.
5. Bousser M-G. Cerebral venous thrombosis: diagnosis and management. J Neurol.
2000;247(4):252–8.
6. Saposnik G, Barinagarrementeria F, Brown RD Jr, et al. American Heart Association Stroke
Council and the Council on Epidemiology and Prevention. Diagnosis and management of
cerebral venous thrombosis: a statement for healthcare professionals from the American Heart
Association/American Stroke Association. Stroke. 2011;42:1158–92.
7. Ferro JM, Canhao P, Bousser MG, Stam J, Barinagarrementeria F, ISCVT Investigators. Early
seizures in cerebral vein and dural sinus thrombosis: risk factors and role of antiepileptics.
Stroke. 2008;39:1152–8.
8. Shulman S, Kearon C, Kakkar AK, et al. Dabigatran versus warfarin in the treatment of acute
venous thromboembolism. N Engl J Med. 2009;361:2342–52.
121
POSTERIOR REVERSIBLE VASOCONSTRICTION SYNDROME
(PRES) AND REVERSIBLE CEREBRAL VASOCONSTRICTION
SYNDROME (RCVS)
Catherine S. W. Albin and Sahar F. Zafar
While PRES and RCVS are not the same disease, there is likely some overlap in the
pathophysiology of the vasoreactivity and endothelial dysfunction that leads to blood
brain barrier breakdown in both [1]. They are also often stroke mimickers. In each,
stopping potentially offending medications and prevention of ischemic and hemor-
rhagic complications are equally important, so they are grouped here for comparison.
124
MANAGEMENT PRINCIPLES IMPORTANT IN BOTH CONDITIONS
□□Review medications and stop offending potentially offending agents; often
requires collaboration with transplant, oncology, or psychiatry team. It is unclear
if medications need to be permanently discontinued or temporarily ceased.
Generally, if there is an equivalent medication, an alternative therapy is tried.
□□ Address any underlying precipitants such as sepsis, acute kidney injury,
(pre)eclampsia
□□ Avoid hypomagnesesmia, aim for high normal range
□□ If excessive hypertension, consider secondary workup (renal artery stenosis,
pheochromocytoma, Cushing’s disease, etc.)
REFERENCES
1. Lee MJ, et al. Blood–brain barrier breakdown in reversible cerebral vasoconstriction syndrome:
implications for pathophysiology and diagnosis. Ann Neurol. 2017;81(3):454–66.
2. Fischer M, Schmutzhard E. Posterior reversible encephalopathy syndrome. J Neurol.
2017;264:1608–16.
3. de Boysson H, Parienti JJ, Mawet J, Arquizan C, Boulouis G, Burcin C, et al. Primary angiitis of
the CNS and reversible cerebral vasoconstriction syndrome: a comparative study. Neurology.
2018;91:e1468–78.
4. Karia SJ, Rykken JB, McKinney ZJ, Zhang L, McKinney AM. Utility and significance of gadolinium-
based contrast enhancement in posterior reversible encephalopathy syndrome. AJNR Am J
Neuroradiol. 2016;37(3):415–22.
125
5. Bartynski WS. Posterior reversible ecenphalopathy syndrome, part 1: fundamental imaging and
clinical features. AJNR Am J Neuroradiol. 2008;29(6):1936–042.
6. Chen SP, Wang SJ. Hyperintense vessels: an early MRI marker of reversible cerebral vasocon-
triction syndrome? Cephalalgia. 2014;34:1038–9.
7. Ansari SB, Rath TJ, Gndhi D. Reversible cerebral vasoconstriction syndromes presenting with
subarachnoid hemorrhage: a case series. J Neurointerv Surg. 2011;3:272–8.
8. Singhal AB, et al. Reversible cerebral vasoconscriction syndromes: analysis of 139 cases. Arch
Neurol. 2011;68(8):1005–12.
9. Miljalski C, Dakay K, Miller-Patterson C, Saad A, Silver B, Khan M. Magnesium for treatment of
reversible cerebral vasoconstriction syndrome. Neurohospitalist. 2016;6(3):111–3.
10. Singhal AB, Topcuoqlu MA. Glucocorticoid-associated worsening in reversible cerebral vasocon-
striction syndrome. Neurology. 2017;88(3):228–36.
126
PART III
Categories of AMS
Drugs/toxins/ Hospital-acquired
Metabolic/systemic medications delirium Primary neruologic
Antiepileptics,
MAOIs, dopamine Infectious:
Infectious: agonists, lithium meningitis,
bacteremia, encephalitis, brain
pneumonia, UTI, abscess
colitis
Steroids, calcineurin
inhibitors
Autoimmune:
paraneoplastic or
Endocrinologic:
other autoimmune
hypo/ hyperglycemia,
encephalitis
hypo/
hyperthyroidism,
hypo/hyperthermia
Neoplastic:
parenchymal brain
mass, dural lesion,
Psychiatric: leptomeningeal
catatonia, carcinomatosis
pseudodementia
130
TESTING TO CONSIDER [1]
Labs For all patients: CBC with differential, BMP, Mg, Phos, LFTs, ABG/VBG, lactate,
troponin, ammonia, UA/UCx, serum/urine tox
To consider by patient history/risk factors: Extended toxicology, ESR, CRP,
BCx, TSH, CK, B12, B1, treponemal Ab, HIV, drug levels for possible offending
home meds, TPO Ab, thyroglobulin Ab
Imaging CTA head & neck if hyperacute-acute change in mental status, and/or focal
neurologic deficits with concern for intracranial hemorrhage or ischemic stroke
MRI brain with contrast if subacute change in mental status and/or focal neurologic
deficits with concern for structural lesion, CNS infection, or above workup without
clear cause
LP (after head Opening pressure (patient should be in lateral decubitus with legs straightened after
imaging) entry into CSF space)
Protein, glucose, cell count & differential, gram stain & culture, HSV 1/2 PCR
If concern for infectious encephalitis: see page 145
If concern for leptomeningeal disease: cytology, flow cytometry, IgH gene
rearrangement, and MYD88 mutation (for CNS lymphomas)
If concern for autoimmune encephalitis: IgG index, oligoclonal bands, autoimmune
encephalopathy panel
EEG Routine EEG if mental status improving, following commands; continuous EEG if
concern for nonconvulsive status epilepticus
MANAGEMENT
Management will primarily depend on etiology. For example, see page 145 for
management of presumed meningitis, 138 for management of seizures, 187 for
elevated ICP, etc.
For Delirium
–– The best treatment is prevention
–– Eliminate precipitating factors, provide frequent reorientation
–– Minimize restraints, lines
–– Promote daytime wakefulness and sleep overnight, minimize nighttime
interruptions
Agitation
–– A Cochrane meta-analysis [2] found that there was no evidence that antipsychot-
ics resolved delirium or altered mortality.
–– However, for severe agitation, atypical antipsychotics like quetiapine are proba-
bly best tolerated and can be considered.
–– Note that given the side effects without demonstrated efficiacy, medications
should not be used when patients can be managed with nonpharmacological
interventions.
131
REFERENCES
1. Douglas VC, Josephson A. Altered mental status. Continuum. 2011;17(5):967–83.
2. Burry L, et al. Antipsychotics for treatment of delirium in hospitalised non-ICU patients. Cochrane
Database Syst Rev. 2018;6(6):CD005594.
132
FRAMEWORK FOR WORKUP OF UNKNOWN BRAIN
“LESION”
Catherine S. W. Albin and Sahar F. Zafar
This is not meant to be an all-inclusive differential for all the pathologies that can
result in brain lesions (meaning FLAIR changes with or without associated post-
gadolinium enhancement), but as a way to take an appropriate history to target the
most likely pathology. Very often a tissue diagnosis is needed, but some lesions
preclude biopsy given location in elegant tissue.
Chart 1: Major categories of unknown lesions. Examples are not all inclusive but
highlight some more common etiologies.
Metastasis
CNS Lymphoma
Adrenoleukodystrophy
Hereditary MELAS
HOST FACTORS
Immune status HIV status? Poorly controlled diabetes? Organ transplant? Review exposure
to any immunomodulators or chemotherapeutic agents, even remote
Exposures Consider location and season. Also inquire about travel, pet/insect/rodent
exposure
Comorbidities Specifically clarify autoimmune history, cancer history, diabetes
Age/sex For example, new onset MS is much more likely in a 30-year-old woman
than a 70-year-old man
Medications/drugs Particularly immunosuppressants and drugs that may result in PRES, PML,
IRIS as well as illicit substances which may put the patient at risk for
endocarditis, vasculopathy, or direct neurologic inflammation
Family history Can provide clues for diseases like leukodystrophies, MELAS, CADASIL
ASSOCIATED SYMPTOMS
Fever Highly concerning for infectious encephalitis or abscesses, but can also be
seen in ADEM, select autoimmune encephalitis cases (NMDA, CASPR2),
and with systemic malignancies
Rashes (or history Rocky Mountain spotted fever, meningococcal meningitis, intravascular
of rash) lymphoma, Bechet’s (oral and genital lesions)
Cough or Sarcoidosis, lung cancer, tuberculosis, aspergillosis, and other fungal
respiratory infections
symptoms
Vision changes Consider demyelinating diseases linked with optic neuritis – NMO, anti-
MOG, MS (see page 175)
All testing should be based on the history and physical. Unless there is a strong
contraindication, all patients should undergo MRI brain with gadolinium
contrast.
Other: Dilated
ophthalmologic
exam (CNS
lymphoma)
135
APPROACH TO FIRST-TIME SEIZURE
Catherine S. W. Albin and Sahar F. Zafar
Remember that not all that shakes or causes a transient spell is a seizure! When
called to evaluate a patient after a first ever “seizure,” remember to keep an open mind
and consider these alternative diagnoses:
□□Transient ischemic attack (particularly limb-shaking TIAs or basilar occlusions
that can result in loss of consciousness)
□□Convulsive syncope (Arrhythmia? PE? Outflow tract obstruction?)
□□Migraine aura or acephalgic migraine
□□Narcolepsy
□□Cerebral Amyloid Angiopathy spells (Transient Focal Neurologic
Episodes (TFNE))
□□Transient global amnesia
□□Psychogenic nonepileptic seizures
□□Panic attack
□□Tremors, rigors, dystonia
□□Fasciculations (particularly with neuromuscular blockers like succinylcholine)
□□Intoxications/withdrawal
CATEGORIES OF FIRST-TIME SEIZURES [1]
REMOTE UNPROVOKED/
ACUTE SYMPTOMATIC SYMPTOMATIC NO CLEAR
PROVOKED SEIZURE SEIZURE SEIZURE ETIOLOGY
Triggered by drugs of Triggered by acute Related to an existing No structural
abuse, toxins, medication- neurologic illnesses such brain lesion – prior lesion, may be
related, acute metabolic as meningitis, traumatic stroke, tumor, remote first presentation
factors (i.e. hyponatremia), brain injury (TBI), or traumatic brain injury of an epilepsy
severe sleep deprivation stroke syndrome
WORKUP
□□History to determine if there have been prior, unrecognized seizures (nocturnal
events? Focal events? Auras?)
□□Review patient’s history about history of meningitis/encephalitis, TBI, strokes,
and prior CNS surgeries
□□Screen patient’s medication lists (clozapine, cephalosporins, fluoroquinolones,
bupropion, tramadol)
Generally accepted that after two unprovoked seizures (spaced apart by >24 h)
AEDs should be initiated as the risk for additional seizures is high (57% at 1 year,
73% by 4 years). However, for a patient with an isolated first seizure, the risk of
future epilepsy needs to be balanced against the risk of AED therapy. Generally, if
just one unprovoked seizure, therapy is only started if there is a high (>60%) likeli-
hood of recurrence.
Factors that raise the risk for future seizures:
–– Prior brain insult or lesions (level A)
–– An EEG with epileptiform abnormalities (level A)
–– Significant neuroimaging abnormality (level B)
–– Nocturnal seizures (level B)
Counseling:
–– Approximately 8–10% of the population experiences 1 seizure; 2–3% go on to
develop epilepsy
–– The risk of recurrence is highest within the first 2 years
–– AEDs each have side effects and risks, most of which are mild and reversible
–– Immediate treatment does not affect the long-term prognosis for epilepsy
** IMPORTANTLY, REMEMBER: Seizures often legally affect patients’ driving
privileges. The duration and extent are state dependent, but patients should be
made aware of these restrictions if they are impacted!**
138
REFERENCES
1. Bergey GK. Management of a first seizure. Continuum (Minneap Minn). 2016;22(1):38–50.
2. Quality Standards Subcommittee of the American Academy of Neurology in cooperation with
American College of Emergency Physicians, American Association of Neurological Surgeons,
and American Society of Neuroradiology. Practice parameter: neuroimaging in the emergency
patient presenting with seizure—summary statement. Neurology. 1996;47(1):288–91.
3. Wellmer J, Quesada CM, Rothe L, et al. Proposal for a magnetic resonance imaging protocol for
the detection of epileptogenic lesions at early outpatient stages. Epilepsia. 2013;54(11):1977–87.
4. Schreiner A, Pohlmann-Eden B. Value of the early electroencephalogram after a first unprovoked
seizure. Clin Electroencephalogr. 2003;34(3):140–4.
5. Gavvala JR, Schuele SU. New-onset seizure in adults and adolescents: a review.
JAMA. 2016;316(24):2657–68.
6. Krumholz A, et al. Evidence-based guideline: management of an unprovoked first seizure
in adults: report of the Guideline Development Subcommittee of the American Academy of
Neurology and the American Epilepsy Society: evidence-based guideline. Epilepsy Curr.
2015;15(3):144–52.
139
PHARMACOLOGY TIPS FOR COMMONLY USED AEDS
Megan E. Barra
FOR STATUS EPILEPTICUS LOADING DOSES AND GUIDANCE, SEE PAGE 259
LEVETIRACETAM
Addressing Hyperammonemia:
142
LACOSAMIDE
DRUG DOSING PK/PD MONITORING CONSIDERATIONS
Lacosamide IV loading T1/2: 13 h EKG, BMP Few drug–drug
(LAC) dose: Metabolism: hepatic interactions. May
Baseline EKG
200–400 mg Elimination: renal require insurance
before initiating
(40% as unchanged prior-authorization.
therapy, watch
Maintenance drug, dose adjust)
for PR interval If CrCl < 30 mL/
dosing: 100 mg Protein binding:
prolongation. Do min: ↓ dose by 75%.
BID PO, may <15%
not use of second HD: ~50% removed.
increase to Bioavailability: 100%
or third degree Add’l dose after HD
200 mg BID Volume of distribution:
AV block or sick session
PO. 0.6 L/kg
sinus syndrome recommended.
IV: PO conversion: 1:1
PHENYTOIN
DRUG DOSING PK/PD MONITORING CONSIDERATIONS
Phenytoin IV loading dose: Michaelis-Menten BMP, CBC w/ diff, Highly protein
(PHT) Use fosphenytoin first-order kinetics at LFTs, albumin, vital bound: low
whenever possible low concentrations, but signs albumin, critical
to load and when zero-order kinetics at illness, uremia, and
Target total level:
phenytoin is being therapeutic drug–drug
10–20 mcg/mL
given IV. concentrations (thus interactions all may
Target free level:
Fosphenytoin at therapeutic affect the
1–2 mcg/mL
results in lower window, slight concentration of
rates of dose changes can active drug.
When loading with
hypotension and cause a dramatic
IV, EKG and BP Calcium and
bradycardia: changes in
monitoring should vitamin D should be
20mg/kg (rec concentration).
be used given risk used in patients on
max 1500mg)
T1/2: 7–42 h of hypotension and chronic therapy.
Correct level for Metabolism: Hepatic bradycardia.
albumin (see Elimination: Nonlinear
Level must be
below). Re-dose hepatic elimination
corrected for low
as needed to (Michaelis-Menten)
albumin.
reach targeted Protein binding: >90%
level (see below). Bioavailability: Variable Long-term usage
(monitor levels when can cause gingival
Daily maintenance
converting from IV to hypertrophy, hair
is ~4–6 mg/kg.
PO) increase, folic acid
100 mg Q8H PO
Volume of distribution: depletion, and
is a reasonable
0.7 L/kg decrease bone
starting dose for
IV: PO conversion: 1:1 density. See AED
most patients.
chart for full side
effect profile
143
Notes on the Dosing of Phenytoin
Correcting for albumin Correct PHT = (Total PHT)/((0.2 × albumin level) + 0.1)
level NOTE: found to be imprecise in critically ill patients
(Winter-Tozer Equation)
Estimating free PHT in Free PHT = 1.69 + 0.139 × (total PHT) − 0.008 (age) − 0.424 (albumin)
critically ill patients + 0.01 (BUN) + 0.288 (critically ill:[yes = 1, no = 0]) [7]
May be more precise in critically ill patients
These equations are NOT valid if the patient is on valproate. Check free levels!
–– Send a free level when possible, but know it may take longer than a total to
result (lab dependent).
–– If a patient is on tube feeds (TF), the TFs must be held for an hour before and
after dosing for oral phenytoin.
When bolusing to Partial loading dose = weight (kg) × 0.7 × ([target PHT level] − [current
achieve a higher level corrected PHT level])
REFERENCES
1. Marion S, et al. Pyridoxine add-on treatment for the control of behavioral adverse effects
induced by levetiracetam in children: a case-control prospective study. Ann Pharmacother.
2018;52(7):645–9.
2. Major P, et al. Pyridoxine supplementation for the treatment of levetiracetam-induced behavior
side effects in children: preliminary results. Epilepsy Behav. 2008;13(3):557–9.
3. Mahmoud A, Tabassum S, Al Enazi S, Lubbad N, Al Wadei A, Al Otaibi A, Jad L, Benini
R. Amelioration of Levetiracetam-Induced Behavioral Side Effects by Pyridoxine. A Randomized
Double Blind Controlled Study. Pediatr Neurol. 2021;119:15–21. PMID: 33823377.
4. Major P, Greenberg E, Khan A, Thiele EA. Pyridoxine supplementation for the treatment of
levetiracetam-induced behavior side effects in children: preliminary results. Epilepsy Behav.
2008;13(3):557–9. PMID: 18647662.
5. Mock CM, Schwetschenau KH. Levocarnitine for valproic-acid-induced hyperammonemic
encephalopathy. Am J Health Syst Pharm. 2012;69(1):35–9. PMID: 22180549.
6. Glatstein M, Bonifacio Rino P, de Pinho S, Scolnik D, Pivko-Levi D, Hoyte C. Levocarnitine for
the Treatment of Valproic Acid-Induced Hyperammonemic Encephalopathy in Children: The
Experience of a Large, Tertiary Care Pediatric Hospital and a Poison Center. PMID: 29232283.
7. Barra ME, Phillips KM, Chung DY, et al. A novel correction equation avoids high-magnitude
errors in interpreting therapeutic drug monitoring of phenytoin among critically ill patients. Ther
Drug Monit 2020;42(4):617–25.
144
APPROACH TO INFECTIOUS ENCEPHALITIS AND MENINGITIS
Catherine S. W. Albin and Megan E. Barra
Encephalitis and meningitis should be considered in any patient with altered mental
status and fever; however, note that septic encephalopathy from a non-CNS source
of an infection is more common than encephalitis.
Encephalitis and meningitis are diagnosed when there is evidence of inflammation in
the brain parenchyma or meninges, respectively. This requires evidence of inflamma-
≥
tion in the CSF (pleocytosis 5 cells/mm3) and/or an abnormality on neuroimaging
(as some immunocompromised hosts or certain infections may not result in signifi-
cant pleocytosis).
EXTRINSIC FACTORS
SIGNS THAT INCREASE CONCERN HOST FACTORS THAT THAT MAY INCREASE
FOR MENINGITIS/ENCEPHALITIS INCREASE RISK FOR CNS RISK FOR CNS
INCLUDE: INFECTION: INFECTION
• Fever • Older age (>65 years old) • S eason (increased
• Headache • T ransplant recipient or on risk for mosquito or
• Nuchal rigidity immunosuppression for tick associated illness
• Altered mental status autoimmune condition in summer)
• Photophobia • A
ctive malignancy and/or • Travel
Patients may also present with: cancer treatment • Geographic origin
• Hydrocephalus • H
IV+, or other acquired/
• Seizures inherited immunodeficiency
• Coma • P rior CNS surgical procedure
• Photophobia • CSF leak
• Nausea/vomiting • R ecent traumatic brain injury
Note that elderly and • External ventricular drain
immunosuppressed patients may have
atypical presentations and no fever or
neck rigidity
146
CNS Biofire FilmArray
Bacterial Pathogens
• Escherichia coli K1,
• Haemophilus influenzae
• Listeria monocytogenes
• Neisseria meningitidis
• Streptococcus agalactiae
• Streptococcus pneumoniae
Viral Pathogens
• Cytomegalovirus (CMV),
• Enterovirus (EV),
• Herpes simplex virus 1 (HSV-1),
• HSV-2,
• Human herpesvirus 6 (HHV-6),
• Human parechovirus (HPeV),
• Varicella-zoster virus (VZV).
Yeast
• Cryptococcus neoformans/Cryptococcus gattii.
Fig. 25.1 MRI T2-weighted image demonstrating dilated Virchow-Robins spaces throughout the
basal ganglia and thalamus in a patient with cryptococcal meningitis. Note that traditioanl antigen
detection is more sensitive than the BioFire Assay for this pathogen
147
TYPICAL CSF FINDINGS IN INFECTIOUS MENINGITIS
TOTAL NUCLEATED CELL COUNT PROTEIN GLUCOSE
Bacterial meningitis 1000–3000, neutrophilic Very Very low. Less than
predominance elevated 2/3 serum glucose.
Often <25 mg/dL
Viral meningitis 200 to >1000, lymphocytic Mildly Normal, although
*HSV encephalitis may have although may be neutrophilic early elevated may be low in
increased red blood cells in in disease some cases
CSF
Fungal meningitis 100–500 Very Low
elevated
TREATMENT
Do not delay starting treatment to get neuroimaging/LP. Draw blood cultures and
then start treatment while arranging for imaging and LP. Most antibiotics result in
CSF sterilization within 4 h; however, Neisseria Meningitidis may be sterilized in
30 min after effective antibiotics.
148
IF CONCERN ATYPICAL
IF HEALTHCARE-ASSOCIATED INFECTIONS (BRUCELLA,
VENTRICULITIS/MENINGITIS (CNS MYCOPLASMA,
DRAINS, NEUROSURGERY, HEAD RICETTSIOSIS, PATIENTS WITH PENICILLIN
TRAUMA) [4]: EHRLICHIOSIS): ALLERGIES:
Use vancomycin + a pseudomonal- Doxycycline 100 mg PO Vancomycin PLUS
active third or fourth-generation or IV Q12H
IgE-mediated reaction (e.g.
cephalosporin (ceftazidime vs.
anaphylaxis, angioedema, hives/
cefepime) or meropenem; remember
urticara):
that these patients are at higher risk
Aztreonam (+TMP/SMX if listeria
for fungal meningitis and infectious
coverage required)
disease guidance is helpful in
Meropenem (has listeria
determining treatment and explant of
coverage)
potentially infected hardware, as
applicable. Severe penicillin allergy (e.g. SJS/
TENs, DRESS, hemolytic anemia):
Fluroquinolone +
TMP/SMX (if indicated for listeria
coverage)
149
Sample Vancomycin Dosing Guidance
CRCL CRCL CRCL <20 ML/
LOADING >80 ML/ 40–80 ML/ CRCL CRCL MIN, AKI, OR
DOSE (ALL MIN AND MIN OR 39–39 20–29 LABILE RENAL
PATIENTS) AGE < 65 AGE > 65 ML/MIN ML/MIN FXN CRRT OR IHD
20–25 15 mg/kg 15 mg/kg 15 mg/kg 15 mg/kg Discuss with Discuss with
mg/kg q8h q12h q24h q24–q48h pharmacy, dose pharmacy, dose
by level by level
Subtherapeutic vancomycin trough before the 4th of 5th dose in patients with stable renal function
First level within 10% of goal Continue same dose with expected accumulation
Level < 5 mcg/mL from goal Increase each dose by 250 mg
Level > 5 mcg/mL lower than Modify dosing interval to next shorter interval (e.g. q12h to q8h)
goal
Supratherapeutic vancomycin trough before the 4th of 5th dose in patients with stable renal function
21–25 mcg/mL Hold next dose until level is expected to be within target then
decrease dose by 250 mg/dose or 500 mg/day
26–30 mcg/mL Hold dose, repeat an interval vancomycin level to inform dosing
>30 mcg/mL Hold dose, re-initiate when an interval level within target range.
Consult Rx for dose recommendations
REFERENCES
1. Venkatesan A, et al. Case definitions, diagnostic algorithms, and priorities in encepha-
litis: consensus statement of the international encephalitis consortium. Clin Infect Dis.
2013;57(8):1114–28.
2. Leber AL, et al. Multicenter evaluation of BioFire FilmArray meningitis/encephalitis panel for
detection of bacteria, viruses, and yeast in cerebrospinal fluid specimens. J Clin Microbiol.
2016;54(9):2251–61.
3. Berkefeld J, Enzensberger W, Lanfermann H. Cryptococcus meningoencephalitis in AIDS:
parenchymal and meningeal forms. Neuroradiology. 1999;41(2):129–33.
4. Tunkel AR, et al. 2017 Infectious Diseases Society of America’s clinical practice guidelines for
healthcare-associated ventriculitis and meningitis. Clin Infect Dis. 2017;64(6):e34–65.
150
NON-INFECTIOUS MENINGITIS
Catherine S. W. Albin and Sahar F. Zafar
Below is a framework and suggested workup for patients with symptoms and signs of
meningitis for whom the entire infectious workup is unrevealing. This is conception-
ally different from “aseptic meningitis” – which is a term that is broadly applied to
meningeal irritation when routine bacterial cultures do not grow. Thus, the term
“aseptic meningitis” includes viral, fungal, spirochete and mycobacterial etiologies
(which do not readily grow in culture); this workup is covered in the previous chapter.
CSF may have a neutrophilic predominance with normal cell glucose. Brain imaging
usually normal.
152
Fig. 26.1 Leptomeningeal Fig. 26.2 Leptomeningeal enhancement
carcinomatosis: T cell lymphoma caking on T1 MR post-contrast in a patient with
the thoracic meninges (arrow) in a neurosarcoidosis who presented
post-gadolinium MRI with rhomboencephalitis
REFERENCES
1. Tattevin P, et al. Aseptic meningitis. Rev Neurol. 2019;175(7–8):475–80.
2. Ungprasert P, Crowson CS, Matteson EL. Characteristics and long-term outcome of neurosar-
coidosis: a population-based study from 1976-2013. Neuroepidemiology. 2017;48(3–4):87–94.
3. Michaël L, Mikaël C, Saskia B, CarolineGiordana, Fanny Burel-Vandenbos, Lydiane M,
JacquesSedat, Denys F, Véronique B, Nihal M, ChristineLebrun-Frenay, Immunoglobulin
G4-related hypertrophic pachymeningitis: A case-oriented review. Neurol Neuroimmunol
Neuroinflamm 2019;6(4):e568.
153
INFLAMMATORY AND AUTOIMMUNE ENCEPHALITIS
Catherine S. W. Albin and Sahar F. Zafar
TYPE EXAMPLE(S)
CNS manifestation Sarcoid, systemic lupus erythematosus
of a systemic disease
Primary autoimmune Steroid responsive encephalitis associated with autoimmune thyroiditis
(STREAT/Hashimoto’s), Miller-Fisher Syndromea, Bickerstaff encephalitis,
Neuromyelitis Optica Spectrum disordersb, multiple sclerosisb, anti-myelin
oligodendrocyte glycoproteinb, Susac’s syndrome
Parainfectious Post-viral cerebellitis, acute demyelinating encephalomyelitis (ADEM)b
Antibody Mediated / See table, page 163
Paraneoplastic
Drug induced Encephalitis associated with interferon-alpha, tumor necrosis factor-alpha,
checkpoint inhibitors and CAR T cells
a
Debated if truly causes encephalitis
b
Primarily demyelinating diseases, covered on 175
4. As mentioned in the chart above, most patients with evidence of inflammation and
a negative infectious workup should be screened with the Autoimmune
Encephalopathy Panel (ARUP/Mayo)
5. Consider utility of advanced neuroimaging and neuro-diagnostics: EEG, Brain
18
FDG-PET, MR Perfusion. These tests may be helpful in establishing CNS inflam-
mation when traditional neuroimaging and CSF sampling fail to demonstrate signifi-
cant inflammation but based on the patient’s clinical symptoms there is still a strong
suspicion of encephalitis.
6. In cases where there is a strong suspicion for an underlying malignancy or primary
inflammatory disorder consider vertex to thigh 18FDG-PET
TREATMENT [3]
For hospitalized patients, immunosuppression may need to begin before a definitive
diagnosis. Ensure that infectious causes of meningitis/encephalitis have been
excluded. To the extent possible, complete imaging and laboratory workup as
156
treatment will impact the yield. This is true of tissue biopsy as well. SAVE
ADDITIONAL CSF AND SERUM PRETREATMENT WHENEVER POSSIBLE.
Fig. 27.1 MRI T1-weighted post gadolinium demonstrating temporal lobe encephalitis in a patient
with post-HSV anti-NMDA-R encephalitis
157
REFERENCES
1. Toledano M, Davies NWS. Infectious encephalitis: mimics and chameleons. Pract Neurol.
2019;19(3):225–37.
2. Rubin DB, et al. Autoimmune encephalitis in critical care: optimizing immunosuppression. Semin
Respir Crit Care Med. 2017;38(06):807–20. Thieme Medical Publishers.
3. Long SS. Encephalitis diagnosis and management in the real world. In: Hot topics in infection
and immunity in children VII, vol. 697. New York: Springer; 2011. p. 153–73.
158
INFECTIOUS WORKUP BY NEUROANATOMICAL
LOCATION: AN ORDERING GUIDE
James Hillis and Catherine S. W. Albin
Can Affect All Patients, but Specific Risk Factors (Rare): Require specific epide-
miological exposures/clinical history. Do not order unless features of the history sup-
port this test.
Fig. 28.1 FLAIR image demonstrating significant atrophy of the temporal lobe in a patient with HSV
limbic encephalitis
Rare EEEa (CSF IgG/ Whipple (CSF EV71 (CSF PCR) JEVe Anaplasmosis &
e
IgM) PCR) JEV EV71 (CSF PCR) Ehrlichiosis (serum PCR
JEVe SLEVe Brucella (serum IgG/ Tick-borne encephalitis and smear)
SLEVe Mumpse IgM) viruse (CSF IgM) Histoplasmosisd (CSF
Powassane Powassane Bechet (see page 155) Polio antigen and IgG/IgM,
EBV (CSF PCR, AHL (see page 155) Rabies urinary antigen) –
serum IgG/IgM) meningitis
Coccidioidesd (CSF fungal
culture, antigen and IgG/
IgM, urinary antigen)
160
Immuno- HHV-6 (CSF PCR) EBV (CSF PCR, CMV (CSF PCR) – Cryptococcusd (CSF
compromised serum IgG/IgM) often causes a cryptococcal
JC virus/PML radiculitis antigen) – meningitis
(CSF PCR) JC virus/PML (CSF
PCR) – white matter
disease
Toxoplasmosis (Serum
Toxo IgG)
ADEM acute disseminated encephalomyelitis, AHL acute hemorrhagic leukoencephalitis, EBV Epstein-Barr virus, EEE eastern equine encepha-
litis, EV71 enterovirus 7, HHV human herpes virus, HSV herpes simplex virus, JC virus/PML John Cunningham virus/progressive multifocal
leukoencephalopathy, JEV Japanese encephalitis virus, LGI-1 leucine-rich glioma-inactivated 1, NMDA-R N-methyl-D-aspartate receptor, NMO
neuromyelitis optica, SLEV St Louis encephalitis virus, TB tuberculosis, VZV varicella zoster virus, WNV West Nile virus
a
Arbovirus Antibody Panel. Usually performed by a state’s viral serology lab. Routinely includes WNV/EEE IgG/IgM with the possibility of adding
further studies through the CDC. You will need to fill out the appropriate form with relevant history. Note that WNV PCR is not sensitive
b
VZV PCR is highly specific but not as sensitive as VZV IgG (compared between serum/CSF). Note that VZV cerebellitis is more common in
children than adults
c
TB CSF PCR, if high index of suspicion
d
Usually cause meningitis, but can cause meningoencephalitis
e
Rare viral encephalitis studies can be ordered through the State Viral Serology Lab (and will then be sent to the CDC). You will need to talk with
the State Lab to organize ordering
161
AUTOIMMUNE ENCEPHALITIS TESTING
Juan Carlos Martinez Gutierrez and James Hillis
ASSOCIATED
ANTIBODY TYPE CANCER(S)A CLINICAL SYMPTOMSB
AChR binding Surface Thymoma Myasthenia gravis
AChR Surface Multiple carcinomas Autonomic dysfunction
ganglionic
AGNA (SOX1) Intracellular Small cell lung cancer Lambert Eaton myasthenic syndrome
AMPA-R Surface Thymoma, lung cancer, Limbic encephalitisc
breast cancer
Amphiphysin Intracellular Breast cancer, small cell Wide clinical spectrum including stiff
lung cancer person syndrome, cerebellar ataxia,
encephalomyelitis
ANNA-1 (Hu) Intracellular Small cell lung cancer, Wide clinical spectrum including sensory
neuroblastoma, thymoma neuropathy, encephalomyelitis, limbic
encephalitis, cerebellar ataxia
ANNA-2 (Ri) Intracellular Small cell lung cancer, Opsoclonus myoclonus, cerebellar ataxia,
breast cancer brainstem encephalitis
ANNA-3 Intracellular Lung cancer Sensory neuropathy, cerebellar ataxia,
encephalomyelitis
164
REFERENCE
1. Linnoila J, Pittock SJ. Autoantibody-associated central nervous system neurologic disorders.
Semin Neurol. 2016;36(04):382–96. Thieme Medical Publishers.
165
APPROACH TO NEW ONSET WEAKNESS
Catherine S. W. Albin and Sahar F. Zafar
The differential diagnosis for a patient with new onset weakness is extremely exten-
sive, but can be significantly narrowed by localizing the weakness and generating a
localization-based differential, which can further be narrowed by the time-course,
patient’s exposures/risk factors, and diagnostic tests [1].
Brain
Myelopathy
Plexopathy
Anterior Horn cells
(“Acute flaccid
myelitis”)
Neuropathy
NMJ
Myopathy
Hyperreflexia.*
Spinal cord
Commonly presents with sensory and motor symptoms.
(Myelopathy)
Look for sensory level.
Physical findings / symptoms suggestive of
Nerve roots Hyporeflexia/areflexia if the nerve root supplies a reflex. LMN fndings.
(Radiculopathy) Radiating pain common.
*In hyperacute setting, the patient may have spinal shock and the reflexes are absent
EVALUATION
Patients with upper motor findings require screening with MRI brain and, as appropri-
ate, C/T spine. The approach to demyelinating diseases is covered on page 175.
Acute onset unilateral pure motor weakness involving face arm and leg is a classi-
cally described lacunar syndrome, and stroke evaluation should proceed (see page
61). Many infectious, inflammatory, neoplastic, and autoimmune causes of encephali-
tis can also cause transverse myelitis. More common causes of myelitis are reviewed
here, but the workup of an unknown lesion in the spinal cord should be undertaken
with the same framework as new brain lesion (see page 133).
168
ELECTED SCREENING EVALUATION FOR INFRATENTORIAL CAUSES OF ACUTE
S
WEAKNESS BY ANATOMICAL SYNDROME
Note that this is not an all-inclusive list of all things that may result in weakness, but a
framework for screening for some of the more common or treatable causes of
weakness that progress quickly enough to warrant hospitalization.
Many etiologies of peripheral weakness progress slowly and are evaluated in the
outpatient setting and are not the focus of this chapter.
Etiologies organized in their category by “VITAMIN” Mnemonic: Vascular, Infectious,
Traumatic, Autoimmune, Metabolic, Iatrogenic, Neoplastic
ANTERIOR HORN
MYELOPATHY [2] CELLS [3] RADICULOPATHY PLEXOPATHY
Causes − Dural AVF Described often as − Lyme disease − Vasculitis
− Stroke “acute flaccid − Acute nerve root − Trauma
− VDRL, VZV, myelitis” compression/trauma − Parsonage-
CMV, EBV, HIV, Infections: − Guillain-Barre (GBS or
−
Turner
Polio
− West Nile Virus
mycoplasma, AIDP) syndrome
169
ANTERIOR HORN
MYELOPATHY [2] CELLS [3] RADICULOPATHY PLEXOPATHY
Possible − MRI C/T spine − MRI C/T spine w/ − MRI C or L spine − Vasculitis
workup w/ and w/o and w/o (localized by exam) w/ screen: ESR/
− dAVF requires − CSF studies (see and w/o; nerve root CRP, ANCA,
MRA-TOFTR page 159 for enhancement commonly C3/C4,
− Demyelinating guidance), seen in GBS hepatitis B/C
workup: see consider Biofire® − CSF routine – look for − Rheumatologic
page 175 − Paraneoplastic albumin-cytologic screen: ANA,
− Viral studies panel from CSF dissociation, consider dsDNA,
from CSF & and serum Biofire®
anti-Ro/La, RF
serum (see − Consider − CSF cytometry and flow − HgbA1c
page 159 for Enterovirus D86 cytology
guidance) testing from − Consider EMG/NCS
− CSF routine respiratory − Lyme (CSF and serum
studies, OCB, specimen, can IgG/IgM)
consider CSF screen with a − Paraneoplastic panel
Biofire® respiratory Biofire® from CSF and serum
− CSF cytology panel for occult − Consider workup for
and flow infections if occult malignancy if
cytometry available high concern for
− Paraneoplastic − Paraneoplastic leptomeningeal
panel from CSF panel (anti-Hu) carcinomatosis
and seruma
− ESR/CRP,
Anti-Ro/La,
ANA/dsDN
− Elements and
Vitamin screen:
B12, vitamin E,
copper, zinc
170
MONONEURITIS NEUROMUSCULAR
MULTIPLEX [8] POLYNEUROPATHY JUNCTION MYOPATHY [6]
Causes − Vasculitis − Vasculitis − Botulism − Crush injury/
(eosinophilic − Lyme disease, HIV, − Myasthenia rhabdomyolysis
granulomatosis leprosy, hepatitis gravis (MG) − Inflammatory
with polyangiitis, C − Lambert Eaton myositis
polyarteritis − Cryoglobulinemia myasthenic (dermatomyositis
nodosa, − Guillain-Barre syndrome and polymyositis)
microscopic Syndrome (AIDP) − Inclusion body
polyangiitis) − AL amyloidosis myositis
− Multiple − Acute intermittent − Necrotizing
compressive lesions porphyria myositis (often
− Systemic − Nutritional/ associated with
rheumatologic diabetic (much HMG-CoA
disease (SLE, more likely to Reductase)
rheumatoid cause sensory − Dystrophies,
arthritis, Sjogren’s neuropathies) metabolic and
most commonly) − Heavy metals mitochondrial
− Diabetes − Critical illness myopathies (not
− Lymphoma, neuropathy covered in workup
Waldenström − Paraneoplastic below as often
macroglobulinemia (anti-Hu/anti-MAG require special
IgM) [5] genetic tests)
− Metabolic, vitamin
deficiency, and
endocrinopathies
− Critical illness
myopathy
− Medication
induced (steroids,
statins,
amiodarone,
colchicine others)
− Cushing syndrome
171
MONONEURITIS NEUROMUSCULAR
MULTIPLEX [8] POLYNEUROPATHY JUNCTION MYOPATHY [6]
Possible − Vasculitis screen: − CSF studies − Anti-AChR − Creatinine kinase
workup ESR/CRP, ANCA, (routine) antibody (CK)
C3/C4, hepatitis − Serum anti-GQ1b − Anti-MuSK − Parathyroid
B/C IgG antibody antibody hormone, TSH
− Rheumatologic (Miller Fisher − Anti-striated − iCal, phosphorous
screen: ANA, variant, see muscle antibody level and basic
dsDNA, anti-Ro/ below) − Paraneoplastic metabolic panel
La, RF − Paraneoplastic panel (VGCC for for hypokalemia
− SPEP/UPEP/IFE panel (anti-Hu) LEMS) − Myositis panel b
172
Selected Variants of Guilain-Barré [7]
Guillain-Barre syndrome = AIDP = Acute inflammatory demyelinating polyneuropathy.
It is actually a polyradiculoneuropathy. There are multiple variants to be aware of
when assessing a patient with weakness.
REFERENCES
1. Berkowitz AL. Clinical neurology and neuroanatomy: a localization-based approach. New York:
The McGraw-Hill Companies; 2017.
2. Kitley JL, et al. The differential diagnosis of longitudinally extensive transverse myelitis. Mult
Scler J. 2012;18(3):271–85.
3. Messacar K, et al. Acute flaccid myelitis: a clinical review of US cases 2012–2015. Ann Neurol.
2016;80(3):326–38.
4. Abdelhady M, Elsotouhy A, Vattoth S. Acute flaccid myelitis in COVID-19. BJR Case Rep.
2020;6(3):20200098.
5. Antoine J-C, Camdessanché J-P. Paraneoplastic neuropathies. Curr Opin Neurol.
2017;30(5):513–20.
6. Katzberg HD, Kassardjian CD. Toxic and endocrine myopathies. Continuum.
2016;22(6):1815–28.
173
7. Dimachkie MM, Barohn RJ. Guillain-Barré syndrome and variants. Neurol Clin.
2013;31(2):491–510.
8. Samson M, et al. Mononeuritis multiplex predicts the need for immunosuppressive or immuno-
modulatory drugs for EGPA, PAN and MPA patients without poor-prognosis factors. Autoimmun
Rev. 2014;13(9):945–53.
174
WORKUP OF NEW DEMYELINATING LESION
Kathryn Holroyd and Kristin Galetta
OVERVIEW
• Demyelinating diseases are autoimmune diseases that damage myelin
in the CNS
• Most present between ages 15–50 years old and are more common in women
• Symptoms include episodes of weakness,
numbness or tingling, monocular vision Clues to multiple sclerosis
loss, and double vision. Cortical symptoms (MS) include that symptoms
such as aphasia or neglect (as seen in worsen in heat (Uhthoff phe-
stroke) are usually not present. nomenon) and patients may
• Symptoms present over hours to days and report episodes of electric
improve in days to months, even resolving shocks down the back of the
completely neck (L’Hermitte sign).
• Neurologic exam findings include asymmet-
ric weakness, sensory loss, eye movement
abnormalities (specifically internuclear ophthalmoplegia), and hyperreflexia
• Most MS is relapse-remitting. However, patients can also present with constant
decline in function over time, indicative of progressive MS (ongoing
neurodegeneration).
• When the primary presenting symptom is severe or bilateral vision loss, a severe
spinal cord syndrome, or intractable nausea/vomiting and hiccups (area postrema
syndrome), neuromyelitis optica (NMO) or anti-myelin oligodendrocyte glycoprotein
(MOG) disease should be considered
• Severe, multifocal deficits with encephalopathy should raise concern for acute
demyelinating encephalomyelitis (ADEM).
DIAGNOSTIC WORKUP
• MRI brain, cervical spine, and thoracic spine
should be ordered w/ and w/out gadolinium. 2017 McDonald Criteria
• If visual loss is concerning for optic neuritis for MS [1]
(ON), MRI orbits w/ gadolinium should also 1. Dissemination in space =
be obtained. evidence of ≥2 lesions in
• Gadolinium enhancement on MRI indicates two separate regions:
active inflammation in a lesion. periventricular, juxtacorti-
• If McDonald criteria are met via imaging and cal, infratentorial, spinal
symptoms, no further testing is needed; MS cord (not optic nerve)
is diagnosed.
ACUTE MANAGEMENT
• If evidence of acute demyelination based on
exam or enhancing lesion on MRI, high-dose Studies to send from CSF:
steroids (1000 mg IV methylprednisolone) –– Total protein
should be administered for 3–5 days. –– Glucose
• Patients with MS exacerbations generally –– White cell count
respond well to IV steroids alone –– Red cell count
• When NMO is suspected based on the –– Gram stain and culture
clinical presentation and imaging, early –– Oligoclonal bands (OCBs)
plasma exchange (PLEX) should be consid- –– IgG Index
ered as limited data indicate this may
improve outcomes [2] Serum studies:
• For patients with ADEM or other demyelinat- –– Anti-aquaporin 4 antibody
ing lesions who do not demonstrate a –– Anti-MOG antibody
clinical response to high-dose steroids, a
treatment course with IVIG (2 g/kg) or
plasma exchange (five sessions) should be
considered, though data is limited [3]
176
DEMYELINATING SYNDROMES
ACUTE
MULTIPLE NEUROMYELITIS ANTI-MOG DEMYELINATING
SCLEROSIS OPTICA SYNDROME ENCEPHALOMYELITIS
Demographics Mean age 28–31 Mean age 31–42 Onset 20–30s More common in
2.3:1 F:M ratio 5:1 F:M ratio Equal M:F ratio children
Low vit D is risk May be ⇑ Considered Often preceded by
factor incidence in those “NMO spectrum infection
⇑ incidence in of African, Latin disorder,” though
northern latitudes American, East may be unique
Asian descent disease
Suggestive Subacute unilateral Severe binocular No findings Acute
exam sensory loss or vision loss specific to encephalopathy with
findings weakness Severe spine anti-MOG focal neurologic
Unilateral vision loss symptoms Can mimic NMO deficits
Double vision Intractable nausea or ADEM
Ataxia or hiccups
Suggestive Ovoid lesions Central spinal No specific Diffuse, bilateral,
imaging Lesions lesions >3 segments findings, but may asymmetric lesions in
findings [4] perpendicular to Bilateral ON have increased white matter of brain
ventricles ON affecting proportion of Multi-segment spine
(“Dawson’s fingers”) chiasm brainstem lesions lesions can be seen
Dorsal spinal lesions Area postrema Can mimic NMO
<2 segments lesion or ADEM
177
REFERENCES
1. Thompson AJ, Banwell BL, Barkhof F, et al. Diagnosis of multiple sclerosis: 2017 revi-
sions of the McDonald criteria. Lancet Neurol. 2018;17(2):162–73. https://doi.org/10.1016/
S1474-4422(17)30470-2.
2. Bonnan M, Valentino R, Debeugny S, et al. Short delay to initiate plasma exchange is the stron-
gest predictor of outcome in severe attacks of NMO spectrum disorders. J Neurol Neurosurg
Psychiatry. 2018;89(4):346–51. https://doi.org/10.1136/jnnp-2017-316286.
3. Keegan M, Pineda AA, McClelland RL, Darby CH, Rodriguez M, Weinshenker BG. Plasma
exchange for severe attacks of CNS demyelination: predictors of response. Neurology.
2002;58(1):143–6. https://doi.org/10.1212/wnl.58.1.143.
4. Tillema JM, Pirko I. Neuroradiological evaluation of demyelinating disease. Ther Adv Neurol
Disord. 2013;6(4):249–68. https://doi.org/10.1177/1756285613478870.
178
APPROACH TO THE “DIZZY” PATIENT
Eric C. Lawson
• A consult for a dizzy patient is one of the most common a neurologist will encounter
• It is important to approach and triage each dizziness consult based on symptom
quality, vascular risk factors, and other common culprits
Timing
• Onset: Acute or
Gradual?
• Episodic, Constant
or Chronic?
TiTraTE
Method
Triggers Targeted
• Positional
• Sound
Exam
• HINTS Exam
• Valsalva
• Cerebellar Signs
• Complex visual
• Dix-Hallpike
stimulation
• Orthostatic Vitals
a plane
l body
Sagitta
45˚
Gravity
Vantage
point
Superior
canal
Posterior
osterior
canal Utriculus
U
Ut
Po
Posterior-canal
P
Gravity aampulla
am
Particles
b
Gravityy
Utriculus
Posterior-canal
ampulla
Superior
canal
Posterior
canal
Vantage
point
Gravity
Particles
180
STEP 3: DIAGNOSING, LOCALIZING, AND TREATING THE CAUSE [3]
DISORDER DURATION TRIGGERS DIAGNOSIS TREATMENT
Benign Acute onset; Turning in bed, Upbeating-torsional Epley Maneuver
paroxysmal duration is reaching upward nystagmus on (see below)
positional seconds Dix-Hallpike
vertigo (BPPV)
Stroke Acute onset; Spontaneous Central nystagmus, Stroke
symptoms last negative head thrust, management (See
days to weeks other neurologic signs page 61)
Posterior Acute onset; Spontaneous Other neurologic TIA management
circulation TIA minutes symptoms
Orthostatic Acute onset; Standing from Postural drop in Hydration,
hypotension seconds sitting or lying blood pressure medication
management
Vasovagal Acute onset; Prolonged Positive tilt table test Hydration
seconds- standing, heat,
minutes stress
Cardiogenic Acute onset; Exertion, heart Arrhythmia, valvular Cardiology
seconds- failure disease Management
minutes
Anxiety Acute or Stress, complex Associated anxiety Anxiety disorder
disorders gradual onset; visual surroundings, and other somatic management
minutes-days crowds symptoms
Vestibular Subacute Spontaneous Unidirectional Consider
neuritis onset; days to horizontal nystagmus. corticosteroids
weeks +Head thrust test
Meniere Subacute Sodium intake Fluctuating hearing Diuretics, restrict
syndrome onset; hours loss sodium intake
Migraine- Gradual onset; Stress, lack of Personal or family hx migraine
associated minutes-days sleep, diet of migraine, other prophylaxis
associated symptoms
Adapted from: Kerber [3]
181
For patients with BPPV, the Epley Maneuver can be tried in which the patients head
is rotated in stages (A) -> (B) -> (C) and then the patient is helped to sit up while
keeping their head facing the downward shoulder until they are completely upright
and then head can be turned midline, as seen in (D). Patients can do these move-
ments at home once trained.
a
Superior Posterior-canal
canal Utriculus ampulla
Gravity
Vantage
point
b
Superior Posterior
canal canal
Vantage
point
Gravity
Particles
Posterior
c canal
Vantage
point
Particles
Gravity
Superior
canal
182
d Superior
Posterior canal
canal
Utriculus
Particles
Posterior-canal
ampulla
REFERENCES
1. Newman-Toker DE, Edlow JA. TiTrATE: a novel, evidence-based approach to diagnosing
acute dizziness and vertigo. Neurol Clin. 2015;33(3):577–99, viii. https://doi.org/10.1016/j.
ncl.2015.04.011. PMID: 26231273; PMCID: PMC4522574.
2. Furman JM, Cass SP. Benign paroxysmal positional vertigo. N Engl J Med. 1999;341(21):
1590–96.
3. Kerber KA, Baloh RW. The evaluation of a patient with dizziness. Neurol Clin Pract.
2011;1(1):24–33. https://doi.org/10.1212/CPJ.0b013e31823d07b6.
183
PART IV
NEUROICU
INTRACRANIAL PRESSURE: THEORY AND MANAGEMENT
STRATEGIES
Melissa Bentley and Catherine S. W. Albin
Monro-Kellie Doctrine
The sum of the volume of brain parenchyma, cerebral spinal fluid, and intracra-
nial blood is constant. An increase in one should cause a decrease in one or
both of the remaining two.
Despite this strong relationship, under normal circumstances, cerebral blood flow
(CBF) can be kept constant over a wide range of cerebral perfusion pressures, and
thus over wide variations in MAPs due to cerebrovascular resistance regulation
(CVR). CVR is an ATP-dependent process that allows the arterioles to vasocon-
strict/dilate.
Note that the Brain Trauma Foundation uses 22 mmHg as its threshold for pathologically elevated ICP.
1
100
80
CBF (mL/100g/min)
60
40
20
0
0 40 80 120 160 200
CPP (mmHg)
However, in vivo, these relationships are more complex. The reasons are multifacto-
rial, but two important concepts need to be considered:
1. Chronically high (or low) MAP resets the “set point” for autoregulation.
(a) For example: chronically hypertensive patients may be able to tolerate much
higher perfusion pressures without hyperemia. But, may have tissue ischemia
even with a CPP of 60–70 mmHg.
188
2. CVR is ATP-dependent. In damaged/ischemic tissue regulation of a constant CBF
for a wide range of CPP is not possible and the relationship between CPP and CBF
becomes more linear.
(a) This means that in some tissues where a CPP of 60 mmHg would have been
adequate for optimal CBF under normal vasoregulation, the vasculature cannot
lower CVR enough to optimize CBF and tissue ischemia results
(b) Or, conversely, a CPP of 120 mmHg, which would be well within most normal
autoregulation capabilities, in damaged brain may result in hyperemia and
reperfusion injury, because of the inability to raise CVR.
100
80
CBF (mL/100g/min)
60
40
20
0
0 40 80 120 160 200
CPP (mmHg)
189
TRATEGIES FOR MONITORING ICP
S
Noninvasive ICP Monitoring
Subarachnoid Externalized
bolt ventricular drain
(EVD)
190
The most common ways of measuring intracranial pressure are with a bolt placed in
the subarachnoid space, an intraparenchymal fiberoptic sensor, or an externalized
ventricular drain (EVD) placed at the intraventricular foramina. Unlike other pressure
sensors, an EVD has the benefit of being able to drain CSF and therefore is not just
a monitoring device but can be used to also treat elevated ICP. For practical tips on
EVD management, see page 197.
When to Monitor
IF TBI [8]: IF NOT TBI [9]:
GCS score ≤8 and CT scan No strict management guidelines.
showing evidence of mass effect In general patients with conditions that would put them at high
risk for developing elevated ICP should be considered for
OR monitoring:
• IVH, especially with early signs of hydrocephalus
When normal CT if:
• Acute hydrocephalus
• Age >40 years
• High-grade (Hunt Hess3–5) subarachnoid hemorrhage
• Motor posturing
• Evidence of shift, herniation, or effacement of basilar cisterns
• Systolic BP <90 mmHg
• Those with clinical signs of increased ICP, see above
• Meningitis/encephalitis when concerned for communicating
hydrocephalus resulting from meningitis and/or or significant
cerebral edema
WAVEFORM INTERPRETATION
Sustained ICPs greater than 20 mmHg are associated with poor outcomes and
should trigger treatment (see page 187). The waveform of the ICP curve is also
helpful in determining brain compliance.
Normal waveform: Waveform when compliance is diminished
P2
20 mmHg 20 mmHg
P1
P3
15 mmHg P1 15 mmHg
P2
P3
1- 4 mmHg
10 mmHg 10 mmHg
5 mmHg
5 mmHg
1 second 1 second
191
P1 = The percussion wave, representing arterial pulsations that are transmitted to
the CSF from the arteries and choroid plexus
P2 = The tidal wave, representing the compliance in the ventricles, thought of as
a rebound of the arterial pulsation
P3 = The dicrotic wave, representing aortic valve closure
When the brain’s compliance is decreased, the P2 wave will be higher than P1. At
this point, very small fluctuations in volume can lead to dramatic increases in
pressure. An intervention to lower ICP can be considered (see page 187).
In addition to beat-to-beat variation, which causes a change of about 1–4 mmHg per
cardiac cycle, there may also be more dramatic changes in the ICP captured by
monitoring. These classic changes were described first by Lunderg in the 1950s and
1960s [10]. The three main categories of ICP variations have been named Lundberg
A, B, and C waves.
Lundberg A Waves
A rapid rise in ICP, continuation on a high level, and then a rapid fall. Also termed
“Plateau Waves.” Clinically may result in changes in consciousness, headache, and
tonic posturing. ICP is dramatically elevated to 50–100 mmHg for 5–20 min before
returning to baseline. Clinically, these waves are driven by a failure of compliance: as
ICP rises, CPP decreases, triggering vasodilation which worsens ICP and edema.
The continued elevation of ICP results in ischemia and failure of cerebral flow to
ischemic territories, ending the positive feedback cycle.
Lundberg B Waves
Variations in the ICP occurring 1/3 to 3 cycles per minute. These fluctuations are on
the order of 5–20 mmHg. Although the phenomenon that causes B waves is complex
and incompletely understood, they likely represent cerebral autoregulation to blood
pressure fluctuations and changes in arterial CO2. These may be seen in normal
individuals as well as those with brain injuries.
Lundberg C Waves
Oscillations 4–8 waves/min associated with variations in the ICP with the respira-
tory cycle.
192
MANAGEMENT OF INCREASED INTRACRANIAL PRESSURE
The most recent editions of the Brain Trauma Foundation (BTF) Guidelines for
Management of Severe Traumatic Brain Injury (sTBI) have removed the algorithms
by which to manage sTBIs due to the lack of evidence of their effectiveness. As a
result, in 2019, 42 physicians gathered together to create the Seattle Severe
Traumatic Brain Injury Consensus Conference (SIBICC) algorithm which provides
three tiers whereby to manage elevated ICPs in severe TBI [8].
Careful consideration should be given when advancing tiers, as higher risk is associ-
ated with the treatment modalities included in the higher tier.
These recommendations were created for severe TBI; however, the principles are
often widely applied to elevated ICP for any cause and guided by Society of
Neurocritical Care Guidelines [11, 12]. Below is an adaptation of sTBI treatment
guidelines that can be applied to all patients with concern for elevated ICP or docu-
mented elevated/refractory ICPs. For further guidance in the management of TBI,
see page 245.
Tier Zero
193
Tier One
• ICP monitoring if meets criteria described above, EVD provides additional ability to drain CSF
• Maintain CPP 60–70 mmHg
For ICP >20 mmHg for 5 min, perform one or more:
• Mannitol or hypertonic saline (HTS) by intermittent bolus for symptoms documented ICP, not
for goal Na/serum Osm goals.
• Increase analgesia & sedation
• Maintain PaCO2 at lower end of normal (35–38 mmHg)
• CSF drainage, if EVD available.
Steroids should NOT be used in any cases other than elevated ICP from vasogenic edema
secondary to tumor or meningitis
Tier Two
• Perform MAP challenge to assess cerebral autoregulation & guide MAP &
CPP goals
• Mild hypocapnia (32–35 mmHg). This is usually only done if there is a plan
for surgical intervention as a prolonged period of hyperventilation may result
in tissue ischemia due to vasocontriction
• Paralytics in adequately sedated patients only if a trial dose is efficacious
Tier Three
• Secondary decompressive craniectomy (see page 200 for data in stroke and
251 for data in TBI)
• Pentobarbital coma, but note significant GI complications and prolonged
sedation with this medication
• Mild hypothermia (35–38 °C)
194
Which Hyperosmolar Therapy to Use?
Both Hypertonic Saline (HTS) and mannitol have benefits and risks. Some compara-
tive studies have found no difference in their effect [13], while a meta-analysis of
small trials found that hypertonic saline is more effective than mannitol in the treat-
ment of elevated ICP [14]. The NCS recommends HTS as first line for
SAH. Generally, the use of which solutions is often an institutional preference and
should be guided by the patient’s personal comorbidities. Both can be used; note with
this strategy careful monitoring of I/Os and electrolyte balance is critically important.
195
Practical Tips for Fever/Shivering Management
Avoid hyperthermia: Avoid shivering:
• Arctic sun or blanketrol for target temp of •S
urface counter-warming w/ Bair Hugger
37.5 °C • Dexmedetomidine infusion
• Central fever management: bromocriptine or • Buspirone 30 mg Q8H
scheduled acetaminophen ×72 h (check LFTs) • Magnesium infusion
• Occasional NSAID administration if bleeding risk • Increase sedation
is low and patient is several days from bleed •P
aralytics (should be tried only when other
(always clear with neurosurgery team). interventions fail)
REFERENCES
1. Aries MJ, Wesselink R, Elting JW, Donnelly J, Czosnyka M, Ercole A, Maurits NM, Smielewski
P. Enhanced visualization of optimal cerebral perfusion pressure over time to support clinical
decision making. Crit Care Med 2016;44(10):e996–9.
2. Sorrentino E, Diedler J, Kasprowicz M, Budohoski KP, Haubrich C, Smielewski P, Outtrim JG,
Manktelow A, Hutchinson PJ, Pickard JD, Menon DK, Czosnyka M. Critical thresholds for cere-
brovascular reactivity after traumatic brain injury. Neurocrit Care. 2012;16(2):258–66.
3. Aries MJ, Czosnyka M, Budohoski KP, Steiner LA, Lavinio A, Kolias AG, Hutchinson PJ, Brady
KM, Menon DK, Pickard JD, Smielewski P. Continuous determination of optimal cerebral perfu-
sion pressure in traumatic brain injury. Crit Care Med. 2012;40(8):2456–63.
4. McNett M, et al. Correlations between hourly pupillometer readings and intracranial pressure
values. J Neurosci Nurs. 2017;49(4):229–34.
5. Chen JW, et al. Pupillary reactivity as an early indicator of increased intracranial pressure: the
introduction of the Neurological Pupil index. Surg Neurol Int. 2011;2:82.
6. Ong C, et al. Effects of osmotic therapy on pupil reactivity: quantification using pupillometry in
critically ill neurologic patients. Neurocrit Care. 2019;30(2):307–15.
7. Rajajee V, et al. Optic nerve ultrasound for the detection of raised intracranial pressure. Neurocrit
Care. 2011;15(3):506–15.
8. Carney N, et al. Guidelines for the management of severe traumatic brain injury. Neurosurgery.
2017;80(1):6–15.
9. Helbok R, Olson DM, Le Roux PD, et al. Intracranial pressure and cerebral perfusion pressure
monitoring in non-TBI patients: special considerations. Neurocrit Care. 2014;21:85–94.
10. Lundberg N, Troupp H, Lorin H. Continuous recording of the ventricular-fluid pressure in patients
with severe acute traumatic brain injury: a preliminary report. J Neurosurg. 1965;22(6):581–90.
11. Cook AM, et al. Guidelines for the acute treatment of cerebral edema in neurocritical care patients.
Neurocrit Care. 2020;32(3):647–66.
12. Stevens RD, Shoykhet M, Cadena R. Emergency neurological life support: intracranial hyperten-
sion and herniation. Neurocrit Care. 2015;23(Suppl 2):S76–82. PMID: 26438459.
13. Francony G, et al. Equimolar doses of mannitol and hypertonic saline in the treatment of increased
intracranial pressure. Crit Care Med. 2008;36(3):795–800.
14. Kamel H, et al. Hypertonic saline versus mannitol for the treatment of elevated intracranial pres-
sure: a meta-analysis of randomized clinical trials. Crit Care Med. 2011;39(3):554–9.
196
MANAGEMENT OF EXTERNAL VENTRICULAR CATHETERS
Catherine S. W. Albin and Sahar F. Zafar
External ventricular drains (EVDs) offer an ability to measure ICP in real time and
also allow for treatment of elevated ICP by drainage of CSF.
EVDs or lumbar drains (LDs) may also be used to treat an active CSF leak post-CNS
procedure or craniofacial trauma, or to prevent a CSF leak after a skull-based
procedure.
EVD SETUP
Often placed at the bedside. The catheter is advanced through a drilled hole in the
skull, preferably through the right frontal lobe and ipsilateral lateral ventricle to the
Foramen of Monro (intraventricular foramen). The catheter has holes for CSF
drainage through the last several centimeters. These holes can be visualized in CT
scans when windowed to the bone window (red arrows).
SELECTED COMPLICATIONS
□□On placement: tract hemorrhage or placement into brain parenchyma
° Most institutions will use a CT scan to confirm placement and rule out
hemorrhage
□□ Ventriculitis
° May be monitored for by routinely sending CSF for glucose/total protein +/-
culture. A dramatic drop in glucose should raise concern for the development
of bacterial infection.
□□ Over-drainage: Can result in subdural hemorrhage, hygromas, upward hernia-
tion, intracranial hypotension, and pneumocephalus
° In most cases, CSF should not be drained more than 20cc/h to prevent this
complication.
198
MALIGNANT MIDDLE CEREBRAL ARTERY INFARCTION
Catherine S. W. Albin and Sahar F. Zafar
CT scan in a young patient with MRI DWI in the same patient CT scan of the same patient
a near-full territory MCA infarct. approximately 30 h post stroke shortly after a post-decompressive
Image approximately 24 h from ictus hemicraniectomy (about 34 h
stroke ictus. after ictus) demonstrating
significant extracranial herniation
from severe cerebral edema.
DHC successfully prevented
transtentorial herniation.
200
MAJOR INCLUSIONS/
TRIAL EXCLUSIONS AND METHODS RESULTS DISCUSSION
HAMLET Enrolled within 96 h from stroke Survival at 91% of patients in the
[3] ictus. Age 18–60. Baseline mRS 12 months was surgical group were
(Lancet 0–1. At least 2/3 territory of MCA 78% in the surgical admitted to the ICU vs.
Neurology territory stroke within 96 h prior to group vs. 41% in 16% in the medical
2009) enrollment. NIHSS ≥ 16 the conservative management group.
nondominant, ≥21 dominant. group.
Attempted to analyze
Gradual decrease in consciousness
25% of both groups quality of life. In the
without confounding factors.
had an mRS 2–3. surgical group 78% of
Conservative treatment included patients had symptoms of
The surgical
osmotherapy, mechanical ventilation mild depression on
group had a
for GCS ≤ 8, ICP monitoring, MADRS metric vs. 58% in
larger portion of
sedation, maintenance of medical group (P = 0.22).
patients with
normothermia, normoglycemia,
mRS of 4–5 In both groups, both
normovolemia.
(53% vs. 15%). patients and caregivers
64 patients. 32 surgical, 32 had very low rates of
conservative treatment. Mean age being dissatisfied with
was 50 (surgical) vs. 47 (medical). treatment ≤10% in all
The mean time from symptom groups. 22 of the 38
onset to randomization was survivors had an mRS
41 (surgical) vs. 45 (medical). score of 4 or 5 at 1 year,
NIHSS was not statistically different and all but one was
(23 vs. 24). happy with the treatment
they received.a
201
MAJOR INCLUSIONS/
TRIAL EXCLUSIONS AND METHODS RESULTS DISCUSSION
DECIMAL Enrolled within 24 h. Age 18–55. Survival at In the craniectomy group,
[4] NIHSS >15. Decreased level of 12 months was 67% of survivors were
(Stroke consciousness. >50% of MCA 75% in surgery vs. home 1 year after
2007) territory. DWI volume >145 cm3. 22% in no surgery. treatment. Age was
Baseline mRS 0–1. Patients could Most patients in the significantly correlated
not have received no surgery group with better outcome in the
tPA. Hemicraniectomy had to be died early (within craniectomy group.
done no more than 6 h after 3.1 ± 1.9 days).
randomization (no more than 30 h
50% of patients in
post-stroke)
the surgical group
Conservative treatment mannitol or achieved an mRS of
furosemide only recommended in 2–3 vs. 22%
hernation, ICP monitoring was not achieving an mRS
recommended; sedation, intubation, of 3 in the
maintenance of normothermia, conservative
normoglycemia, normovolemia were treatment group.
at the discretion of treating
physician.
38 patients. 20 surgical, 18
conservative. Mean age/NIHSS
was 43.5/22.5 (surgical) vs.
43.3/23.4 (medical).
202
MAJOR INCLUSIONS/
TRIAL EXCLUSIONS AND METHODS RESULTS DISCUSSION
DESTINY II Enrollees were >60 years old. Survival at After 12 months, 6% of
[5] Enrolled within 48 h of stroke ictus. 12 months was patients >60 had an mRS
(NEJM All were treated in the ICU. NIHSS 57% in the surgical of 3, compared to 43% of
2014) >14 (nondominant), >19 group vs. 24% in younger patients.
(dominant). Decreased level of the medical group.
Early hemicraniectomy
conscionssness. >2/3 of the MCA
6% surgical and 5% significantly increased
territory and at least a portion of
medical achieved an probability of survival but
basal ganglia. Baseline mRS 0–1.
mRS 3. None <3. most survivors had
Conservative treatment: ICU substantial disability.
The surgical
treatment for all patients. At
group had a
physicians’ discretion: osmotherapy,
larger portion of
mechanical ventilation for GCS ≤ 8,
patients with an
ICP monitoring, sedation,
mRS 4–5 (51%
maintenance of normothermia,
vs. 19%). mRS 5
normoglycemia, normovolemia.
was more than
112 patients included. Mean age 2× more
was 70 (range 61–82). Average common in the
NIHSS 20 (surgical) vs. 21 surgery group
(conservative). Mean time from (28% vs. 13%)
onset of symptoms to
hemicraniectomy = 28 h.
a
This is an interesting finding, but as noted in the trial, should be interpreted with caution as the
patient feeling compelled to give the desired answer cannot be excluded as the question was
not predefined
Notes:
The pooled analysis [6] of DESTINY I, HAMLET, and DECIMAL demonstrated that
early hemicraniectomy (w/in 48 h):
–– Increased 1 year survival from 29% to 78%
–– Resulted in a low rate of severe disability (mRS 5): 4%
–– Resulted in a moderate percentage of patients with a good outcome (mRS 2–3):
43% vs. 21%
–– Number needed to treat to obtain a survival of mRS ≤ 3: 4
A recent meta-analysis reviewing seven trials [7] found that there was no evidence of
heterogeneity of treatment outcome based on the presence of aphasia, stroke
severity, age, and involvement of other vascular territories in addition to the MCA. In
regard to age, the positive effect of surgery was smaller and the Confidence Interval
crossed 1. In this meta-analysis, there was a slight favoring of medical treatment for
patients being treated after 48 h.
203
REFERENCES
1. Hacke W, et al. Malignant middle cerebral artery territory infarction: clinical course and prognos-
tic signs. Arch Neurol. 1996;53(4):309–15.
2. Jüttler E, et al. Decompressive surgery for the treatment of malignant infarction of the middle
cerebral artery (DESTINY) a randomized, controlled trial. Stroke. 2007;38(9):2518–25.
3. Hofmeijer J, et al. Surgical decompression for space-occupying cerebral infarction (the
Hemicraniectomy After Middle Cerebral Artery infarction with Life-threatening Edema Trial
[HAMLET]): a multicentre, open, randomised trial. Lancet Neurol. 2009;8(4):326–33.
4. Vahedi K, et al. Sequential-design, multicenter, randomized, controlled trial of early decom-
pressive craniectomy in malignant middle cerebral artery infarction (DECIMAL trial). Stroke.
2007;38(9):2506–17.
5. Jüttler E, et al. Hemicraniectomy in older patients with extensive middle-cerebral-artery stroke. N
Engl J Med. 2014;370(12):1091–100.
6. Vahedi K, Hofmeijer J, Juettler E, et al. Early decompressive surgery in malignant middle
cerebral artery infarction: a pooled analysis of three randomised controlled trials. Lancet Neurol.
2007;6:215–22.
7. Reinink H, et al. Surgical decompression for space-occupying hemispheric infarction: a sys-
tematic review and individual patient meta-analysis of randomized clinical trials. JAMA Neurol.
2021;78(2):208–16.
204
INTRAPARENCHYMAL HEMORRHAGE
Catherine S. W. Albin and Sahar F. Zafar
Intraacerebral hemorrhage
Hemorrhagic metastasis
Hemorrhagic
(frequently @ gray-white
metastasis
junction)
Endocarditis (septic
emboli)
Hypertension
CT scan demonstrating a basal SWI MRI demonstrating multiple CT scan of patient with cortical
ganglia hemorrhage secondary features of cerebral amyloid venous sinus thrombosis. Note
to long-standing poorly angiopathy: cortical bleeds, the cortical location with
controlled hypertension microhemorrhages, and surrounding hypodensity
superficial siderosis (representing a combination of
vasogenic and cytogenic edema)
CT scan in patient with mitral MRI T2 FLAIR of hemorrhagic CT scan demonstrating pontine
valve endocarditis and multiple cerebellar metastasis hemorrhage secondary to
mycotic aneurysms. Bleed likely long-standing poorly controlled
due to hemorrhagic hypertension
transformation of septic embolic
infarct
206
EARLY MANAGEMENT [1]
□□Somnolence common, assess need for
intubation (GCS ≦8) For tPA-Related Bleeding:
□□ Consider need for external ventricular
□□
Hold tPA
drain, hemicraniectomy, or other
surgical intervention in discussion with
□□
STAT CBC, PT, PTT,
fibrinogen, and D-dimer
□□
neurosurgery – see Trials, page 211
Confirm SBP < 140 vs 1601 – see
□□
Type and Screen and if
systemic bleeding,
Trials, page 211
□□ Confirm received reversal of anticoagu-
cross match
□□
Once ICH confirmed,
lation, if applicable (see page 215)
□□ Review head CT
see page 215 for
□□ Review or order CT angiogram based
reversal strategies
on potential etiology, review images to
look for a “spot sign” (Fig. 36.1) or a vascular malformation
□□ Confirm that the patient is ordered for stability imaging2
□□ Consider if a conventional angiogram should be pursued (young/atypical
pattern/unexplained bleed)
□□ Consider need for CT venogram vs MRI w/ and w/o + MR venogram if high
concern for venous sinus thrombosis
□□ Review EKG
207
SCORING SYSTEMS [3–5]
208
Boston Criteria for Cerebral Amyloid Angiopathy:
Definite cerebral amyloid angiopathy:
Full post-mortem examination reveals lobar, cortical, or cortical/subcortical
hemorrhage and pathological evidence of severe CAA
Probable cerebral amyloid angiopathy (w/ pathological evidence):
Clinical data and pathological tissue demonstrate a hemorrhage and some
degree of vascular amyloid deposition. Does not have to be post-mortem.
Probable cerebral amyloid angiopathy:
Pathological confirmation not required
• Patient older than 55 years
• Appropriate clinical history
• MRI findings demonstrate multiple hemorrhages of varying sizes/ages with no
other explanation
Possible cerebral amyloid angiopathy:
• Patient older than 55 years
• Appropriate clinical history
• MRI findings reveal a single lobar, cortical, or cortical/subcortical hemor-
rhage w/o another cause, multiple hemorrhages with a possible other cause,
or some hemorrhage in an atypical location
209
Fig. 36.1 A “Spot Sign” – the appearance of contrast density within the hematoma bed which is a
concern for ongoing active extravasation of contrast – is seen here in a patient with a large basal
ganglia hemorrhage
REFERENCES
1. Hemphill JC III, et al. Guidelines for the management of spontaneous intracerebral hemorrhage:
a guideline for healthcare professionals from the American Heart Association/American Stroke
Association. Stroke. 2015;46(7):2032–60.
2. Wada R, et al. CT angiography “spot sign” predicts hematoma expansion in acute intracerebral
hemorrhage. Stroke. 2007;38(4):1257–62.
3. Hemphill JC, et al. The ICH score. Stroke. 2001;32(4):891–7.
4. Rost NS, et al. Prediction of functional outcome in patients with primary intracerebral hemor-
rhage: the FUNC score. Stroke. 2008;39(8):2304–9.
5. Knudsen KA, et al. Clinical diagnosis of cerebral amyloid angiopathy: validation of the Boston
criteria. Neurology. 2001;56(4):537–9.
6. Becker KJ, Baxter AB, Cohen WA, Bybee HM, Tirschwell DL, Newell DW, Winn HR, Longstreth
WT Jr. Withdrawal of support in intracerebral hemorrhage may lead to self-fulfilling prophecies.
Neurology. 2001;56(6):766–72. https://doi.org/10.1212/wnl.56.6.766. PMID: 11274312.
210
INTRACRANIAL HEMORRHAGE – LANDMARK TRIALS
Catherine S. W. Albin and Sahar F. Zafar
Goal was reduction in death or disability At 1 h, the mean systolic blood
(mRS 3–6) at 90 days. 2839 patients, 1403 pressure was 150mmHg in the
to early intensive treatment, 1436 to usual intensive treatment group. (33%
care. The mean interval between symptom achieved the target <140) vs.
onset and randomization was ~4 h. 164mmHg in the standard treatment
group. Primary treatment failure was
seen in 66% of the participants within
1 h after randomization.
212
TRIAL TRIAL DESIGN MAJOR FINDINGS
CLEAR Randomized, multicenter, double-blind, At 180 days the treatment group had
IVH-III placebo-controlled. Patients with ICH <30 cc lower mortality 18% vs 29%;
(Lancet 2017) with IVH obstructing the third or fourth however more patients with severe
[5] ventricle. Patients were screened for EVD disability (17% vs. 9%). Rates of
tract hemorrhage and if none present at symptomatic bleeding, ventriculitis
6–12 h post-placement, patients were and serious adverse events were not
randomized to received rtPA vs. normal statistically different.
saline infusions. Excluded if coagulopathy or
Failed to improved outcomes to the
confirmed/suspicion of aneurysm, AVM,
cutoff of mRS 3 at 180 days, but rtPA
other vasc malformation.
did not have appear to have
Treatment was up to 12 doses of 1 mg significant adverse events.
alteplase q8h. CT scans were obtained every
Subgroup analysis failed to show any
24 h.
treatment benefit in patients with
Goal was improved outcome (mRS < 4) at thalamic hemorrhage or those with
180 days. 500 patients included. 249 IVH < 20 cc
alteplase vs. 251 saline.
CT scan on day 1 from a patient who was then CT scan of the same patient on day 5 which
treated with eight doses of intrathecal tPA per demonstrates significant reduction in IVH and
CLEAR trial guidelines (Q8H doses after hydrocephalus.
stabilization of bleeding and no evidence of EVD
tract hemorrhage).
213
REFERENCES
1. Anderson CS, et al. Rapid blood-pressure lowering in patients with acute intracerebral hemor-
rhage. N Engl J Med. 2015;368(25):2355–65.
2. Qureshi AI, et al. Intensive blood-pressure lowering in patients with acute cerebral hemorrhage.
N Engl J Med. 2016;375(11):1033–43.
3. Mendelow AD, et al. Early surgery versus initial conservative treatment in patients with spon-
taneous supratentorial lobar intracerebral haematomas (STICH II): a randomised trial. Lancet.
2013;382(9890):397–408.
4. Hanley DF, et al. Efficacy and safety of minimally invasive surgery with thrombolysis in intra-
cerebral haemorrhage evacuation (MISTIE III): a randomised, controlled, open-label, blinded
endpoint phase 3 trial. Lancet. 2019;393(10175):1021–32.
5. Hanley DF, et al. Thrombolytic removal of intraventricular haemorrhage in treatment of severe
stroke: results of the randomised, multicentre, multiregion, placebo-controlled CLEAR III trial.
Lancet. 2017;389(10069):603–11.
214
REVERSAL OF SELECTED ANTITHROMBOTICS
Catherine S. W. Albin and Megan E. Barra
216
Dabigatran First line: Idaracizumab Baseline STAT aPTT, 2–4 h, and 12–24 h
(Praxbind): 5 g IV administered post administration dTT or TT may be
as two sequential 2.5 g IV infusions helpful in quantifying presence of clinically
no more than 15 min apart significant dabigatran levels in patients with
unknown last administration time or
If Idaracizumab unavailable
ongoing hemorrhage, if readily available
4F-PCC or aPCC: 50 IU/kg (max
dose 5000 IU) In clinical trials, a small proportion of
patients were found to have rebound of
Reversal should be done if the last
dabigatran levels ≥12 h after
dose was administered within 3–5
administration, which was associated with
half-lives (e.g. past 2–4 days).
recurrent or continuous bleeding. This is
Longer if moderate-severe renal
due to re-distribution from adipose tissue
insufficiency. Elevated TT or dTT
after reversal of dabigatran. Patients with
can signify presence of clinically
renal dysfunction at highest risk for
significant dabigatran levels if
phenomenon. May consider re-dosing
unknown last administration
idarucizumab if ongoing hemorrhage or
need for emergent procedures with
abnormal coagulation labs [17].
Apixaban, Andexanet alfa: Baseline anti-Xa for UFH and LMWH may
rivaroxaban Indicated for reversal of apixaban be helpful in ruling out presence of
or rivaroxaban if last dose within clinically significant rivaroxaban- or
previous 18 h: apixaban-levels if undetectable in patients
with unknown last administration time.
Time since last dose
FXa inhibitor Last dose
<8 h or
Or, baseline DOAC-specific anti-Xa levels if
8 –18 h
<10 mg
unknown
Low dosea
readily available
Rivaroxaban >10 mg or
High doseb
unknown
Low dosea Andexanet alfa:
<5 mg Low dosea
Apixaban >5 mg or
High doseb
Short duration of action. Rebound
unknown
a
Andexanet alfa low dose regimen: initial IV bolus 400 mg anticoagulation observed within 2-h of
infused at a target rate of 30 mg/min followed by a 4 mg/min
continuous intravenous infusion for up to 120 min cessation of IV infusion to levels observed in
b
Andexanet alfa high dose regimen: initial IV bolus 800 mg
infused at a target rate of 30 mg/min followed by a 8 mg/min patients not reversed with andexanet alfa.
continuous intravenous infusion for up to 120 min
Best used in patients with acute
hemorrhages at high risk for hematoma
Alternative: 4F-PCC 25–50
expansion.
IU/kg (max dose 5000 IU)
4F-PCC:
No standardized dosing available, most
guidelines recommend 50 IU/kg, though
lower dose regimens (25–37.5 IU/kg) have
been increasingly reported with similar
hemostatic effectiveness rates
217
Edoxaban Andexanet alfa: High-dose Baseline anti-Xa for UFH and LMWH may
regimen (off-label) be helpful in ruling out presence of
Initial IV bolus 800 mg infused at a clinically significant rivaroxaban or
target rate of 30 mg/min followed apixabanlevels if undetectable in patients
by a 8 mg/min continuous with unknown last administration time.
intravenous infusion for up to 120
Edoxaban underrepresented in studies
Alternative: 4F-PCC 25–50 evaluating safety and efficacy of andexanet
IU/kg alfa. High-dose regimen is recommended.
Parenteral anticoagulants [1]
Unfractionated Protamine 1 mg/100 units of Re-check STAT aPTT 15 min after protamine
heparin heparin administered in past 3 h, administration, if aPTT remains elevated,
max single dose 50 mg repeat 0.5 mg/100 units of heparin
administered in previous 3 h.
Reversal for prophylactic dosing not
recommended
Enoxaparin Last enoxaparin Re-check STAT aPTT 2–4 h after protamine
administration Protamine Dose
1 mg protamine per 1 mg
administration. If aPTT remains elevated or
<8 h
enoxaparin*
ongoing significant bleeding, repeat
0.5 mg protamine per 1 mg
>8 h
enoxaparin* 0.5 mg/1 mg enoxaparin.
*Max single dose 50 mg
Protamine does not completely neutralize
anti-Xa activity (maximum neutralization
60–75%).
Reversal for prophylactic dosing not
recommended
Fondaparinux aPCC: 20 IU/kg Reversal for prophylactic dosing not
If unavailable: rFVIIa: 90 mcg/kg recommended
×1
218
Fibrinolytics [18]
Recombinant Stop infusion immediately. Reverse Baseline STAT Fibrinogen *do not wait for
tissue if within 24 h of rtPA infusion. lab to result to administer cryoprecipitate*
plasminogen Repeat fibrinogen 30 min- 1 h after
Cryoprecipitate: Potential benefit
activator (rtPA) administration and every 2 h until bleeding
in all patients
controlled
• Empiric administration of
10 units in all patients
• Repeat dose until fibrinogen
≥150 mg/dL
REFERENCES
1. Win JJ, Rabinstein AA, et al. Guideline for reversal of antithrombotics in intracranial hemorrhage.
Neurocrit Care. 2016;24:6–46.
2. Jessica M, et al. Effect of desmopressin acetate on acute spontaneous intracranial hemorrhage
in patients on antiplatelet therapy. Journal of the Neurological Sciences. 2022:120142.
3. Baharoglu MI, Cordonnier C, Salman RA, et al. Platelet transfusion versus standard care after
acute stroke due to spontaneous cerebral hemorrhage associated with antiplatelet therapy
(PATCH): a randomised, open-label, phase 3 trial. Lancet. 2016;387(10038):2605–13.
4. Lip GYH, Banerjee A, Boriani G, et al. Antithrombotic therapy for atrial fibrillation: CHEST guide-
line and expert panel report. Chest. 2018;154(5):1121–201.
5. Witt DM, Nieuwlaat R, Clark NP, et al. American Society of Hematology 2018 guidelines for man-
agement of venous thromboembolism: optimal management of anticoagulation therapy. Blood
Adv. 2018;2(22):3257–91.
6. Tomaselli GF, Mahaffey KW, Cuker A, et al. 2020 ACC expert consensus decision pathway on
management of bleeding in patients on oral anticoagulants: a report of the American College of
Cardiology Solution Set Oversight Committee. J Am Coll Cardiol. 2020;76(5):594–622.
7. January CT, Wann LS, Calkins H, et al. 2019 AHA/ACC/HRS focused update of the 2014 AHA/
ACC/HRS guideline for the management of patients with atrial fibrillation: a report of the American
College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines
219
and the Heart Rhythm Society in collaboration with the Society of Thoracic Surgeons. Circulation.
2019;140(2):e125–51.
8. Christensen H, Cordonnier C, Kõrv J, et al. European Stroke Organisation guideline on reversal
of oral anticoagulants in acute intracerebral haemorrhage. Eur Stroke J. 2019;4(4):294–306.
9. Varga C, Al-Touri S, Papadoukakis S, Caplan S, Kahn S, Blostein M. The effectiveness and safety
of fixed low-dose prothrombin complex concentrates in patients requiring urgent reversal of war-
farin. Transfusion. 2013;53(7):1451–8.
10. Klein L, Peters J, Miner J, Gorlin J. Evaluation of fixed dose 4-factor prothrombin complex. con-
centrate for emergent warfarin reversal. Am J Emerg Med. 2015;33(9):1213–8.
11. Hirri HM, Green PJ. Audit of warfarin reversal using a new Octaplex reduced dose protocol.
Transfus Apher Sci. 2014;51(2):141–5.
12. Khorsand N, Veeger NJ, van Hest RM, Ypma PF, Heidt J, Meijer K. An observational, prospec-
tive, two-cohort comparison of a fixed versus variable dosing strategy of prothrombin complex
concentrate to counteract vitamin K antagonists in 240 bleeding emergencies. Haematologica.
2012;97(10):1501–6.
13. Khorsand N, Veeger NJ, Muller M, et al. Fixed versus variable dose of prothrombin complex con-
centrate for counteracting vitamin K antagonist therapy. Transfus Med. 2011;21(2):116–23.
14. Abdoellakhan RA, Miah IP, Khorsand N, Meijer K, Jellema K. Fixed versus variable dosing of
prothrombin complex concentrate in vitamin K antagonist-related intracranial hemorrhage: a ret-
rospective analysis. Neurocrit Care. 2017;26(1):64–9.
15. Goldstein JN, Refaai MA, Milling TJ, et al. Four-factor prothrombin complex concentrate ver-
sus plasma for rapid vitamin K antagonist reversal in patients needing urgent surgical or
invasive interventions: a phase 3b, open-label, non-inferiority, randomised trial. Lancet.
2015;385(9982):2077–87.
16. Sarode R, Milling TJ, Refaai MA, et al. Efficacy and safety of a 4-factor prothrombin complex
concentrate in patients on vitamin K antagonists presenting with major bleeding: a randomized,
plasma-controlled, phase IIIb study. Circulation. 2013;128(11):1234–43.
17. Pollack CV, Reilly PA, van Ryn J, et al. Idarucizumab for dabigatran reversal—full cohort analysis.
N Engl J Med. 2017;377(5):431–41.
18. Yaghi S, Willey JZ, Cucchiara B, et al. Treatment and outcome of hemorrhagic transformation
after intravenous alteplase in acute stroke: a scientific statement for healthcare professionals
from the American Heart Association/American Stroke Association. Stroke. 2017;48:e343–61.
220
AN IN-DEPTH REVIEW OF REVERSING DIRECT FACTOR
XA-INHIBITOR-RELATED HEMORRHAGES
Megan E. Barra
BACKGROUND
• In landmark clinical trials for DOAC therapy, incidence of ICH was 0.51% and
ICH-mortality ranged from 45.3% (apixaban) to 48% (rivaroxaban) before reversal
agents were available [1, 2].
• Withholding reversal in DOAC-associated ICH has been associated with 1.5-fold
increased risk of death and worse functional outcomes [3].
• Mechanism of action: binds to and sequesters Factor-Xa inhibitors with high affinity.
Also found to inhibit activity of tissue factor pathway inhibitor (TFPI) resulting in an
increase in tissue factor-initiated thrombin generation.
222
The decision about which agent (andexanet alfa vs PCC) for Factor Xa inhibitor
reversal is often guided by which agent is available on hospital formulary.
REFERENCES
1. Held C, Hylek EM, Alexander JH, et al. Clinical outcomes and management associated with
major bleeding in patients with atrial fibrillation treated with apixaban or warfarin: insights from the
ARISTOTLE trial. Eur Heart J. 2015;36(20):1264–72.
2. Piccini JP, Garg J, Patel MR, ROCKET AF Investigators, et al. Management of major bleeding
events in patients treated with rivaroxaban vs. warfarin: results from the ROCKET AF trial. Eur
Heart J. 2014;35(28):1873–80.
3. Apostolaki-Hansson T, Ullberg T, Pihlsgard M, Norrving B, Petersson J. Reversal treatment
in oral anticoagulant-related intracerebral hemorrhage-an observational study based on the
Swedish stroke register. Front Neurol. 2020;11:760.
4. Connolly SJ, Crowther M, Eikelboom JW, et al. ANNEXA-4 investigators. Full study report of andex-
anet alfa for bleeding associated with factor Xa inhibitors. N Engl J Med. 2019;380(14):1326–35.
5. Barco S, Whitney CY, Coppen M, et al. In vivo reversal of the anticoagulant effect of rivaroxaban
with four-factor prothrombin complex concentrate. Br J Haematol. 2016;172(2):255–61.
6. Eerenberg ES, Kamphuisen PW, Sijpkens MK, et al. Reversal of rivaroxaban and dabigatran by
prothrombin complex concentrate: a randomized, placebo-controlled, crossover study in healthy
subjects. Circulation. 2011;124(14):1573–9.
7. Majeed A, Ågren A, Holmström M, et al. Management of rivaroxaban- or Apixaban-
associated major bleeding with prothrombin complex concentrates: a cohort study. Blood.
2017;130(15):1706–12.
8. Gerner ST, Kuramatsu JB, Sembill JA, et al. Association of prothrombin complex concentrate
administration and hematoma enlargement in non-vitamin K antagonist oral anticoagulant-related
intracerebral hemorrhage. Ann Neurol. 2018;83(1):186–96.
9. Schulman S, Gross PL, Ritchie B, et al. Prothrombin complex concentrate for major bleeding on
factor Xa inhibitors: a prospective cohort study. Thromb Haemost. 2018;118(5):842–51.
10. Wilsey HA, Bailey AM, Schadler A, et al. Comparison of low- versus high-dose four-factor pro-
thrombin complex concentrate (4F-PCC) for factor Xa inhibitor-associated bleeding: a retrospec-
tive study. J Intensive Care Med. 2020;36(5):597–603. [epub ahead of print]
11. Castillo R, Chan A, Atallah S, et al. Treatment of adults with intracranial hemorrhage on apixa-
ban or rivaroxaban with prothrombin complex concentrate products. J Thromb Thrombolysis.
2021;51(1):151–8.
12. Lipari L, Yang S, Milligan B, et al. Emergent reversal of oral factor Xa inhibitors with four-factor
prothrombin complex concentrate. Am J Emerg Med. 2020;38(12):2641–5.
13. Panos NG, Cook AM, John S, et al. Factor Xa inhibitor-related intracranial hemorrhage: results
from a multicenter, observational cohort receiving prothrombin complex concentrates. Circulation.
2020;141(21):1681–9.
223
INTRACRANIAL HEMORRHAGE – MANAGEMENT
OF ANTICOAGULATION
Juan Carlos Martinez Gutierrez
EARLY MANAGEMENT
Hematoma expansions occur in 14–38%, most happen in first hour (26%) and first
day (12%) and rarely in subsequent 2 weeks (<2%).
Factors associated with expansion [1].
–– Liver dysfunction (18.3% higher incidence).
–– Spot sign (77% expand when present, 96% do not expand when absent) [2].
–– Hematoma volume <20 cc (8.2%) vs. >20 cc (31.5%).
–– Irregular shape (13.2% higher incidence).
–– Antithrombotics.
All antithrombotics should be held and reversed (if applicable) in the first
24 hrs (see page 215 for reversal agents).
DVT Prevention:
□□
Start pneumoboots on admission. DVTs can occur in 17–40.4% of ICH patients
without prophylaxis and only 6.7% with boots [3, 4].
□□Chemical DVT prophylaxis can be started 24–48 after bleed stability is con-
firmed, if there are no other contraindications (elevated INR, low plts, etc.).
TIMING OF RESUMPTION
Still undefined. In one study of 59 patients that restarted anticoagulation after ICH,
the combined risk of recurrent intracranial hemorrhage or ischemic stroke reached a
nadir if warfarin was resumed after approximately 10–30 weeks [18].
Clinical practice varies though, and many experts would resume anticoagulation at
1–2 months post-bleed depending on the reason to need anticoagulation.
226
Take Away Recommendations for Management
–– Whenever possible, obtain MRI to assess for CMB, cSS, and leukoaraiosis
to help risk-stratify.
–– Whenever possible, consider devices that would limit the need for life-long
pharmacologic anticoagulation such as a WATCHMAN device (AF) or IVC
filter (VTE).
–– For hypertensive/deep hemorrhages, restarting anticoagulation when nec-
essary can be considered; DOACs are preferable.
–– In patients with suspected CAA, anticoagulation is usually not resumed for
atrial fibrillation; PE and DVTs must be evaluated on a case-by-case sce-
nario. If needed DOAC use is likely preferable.
REFERENCES
1. Fujii Y, et al. Hematoma enlargement in spontaneous intracerebral hemorrhage. J Neurosurg.
2009;80:51–7.
2. Wada R, et al. CT angiography ‘spot sign’ predicts hematoma expansion in acute intracerebral
hemorrhage. Stroke. 2007;38:1257–62.
3. Ogata T, et al. Deep venous thrombosis after acute intracerebral hemorrhage. J Neurol Sci.
2008;272:83–6.
4. Dennis M. Effectiveness of intermittent pneumatic compression in reduction of risk of deep vein
thrombosis in patients who have had a stroke (CLOTS 3): a multicentre randomised controlled
trial. Lancet. 2013;382:516–24.
5. Bae H, et al. Recurrence of bleeding in patients with hypertensive intracerebral hemorrhage.
Cerebrovasc Dis. 1999;9:102–8.
6. Vermeer SE, Algra A, Franke CL, Koudstaal PJ, Rinkel GJE. Long-term prognosis after recovery
from primary intracerebral hemorrhage. Neurology. 2002;59:205–9.
7. Biffi A, et al. Oral anticoagulation and functional outcome after intracerebral hemorrhage. Ann
Neurol. 2017;82:755–65.
8. Hemphill JC III, Greenberg SM, Anderson CS, et al. Guidelines for the management of spontane-
ous intracerebral hemorrhage: a guideline for healthcare professionals from the American Heart
Association/American Stroke Association. Stroke. 2015;46:2032–60.
9. Eckman MH, Rosand J, Knudsen KA, Singer DE, Greenberg SM. Can patients be anticoagulated
after intracerebral haemorrhage? A decision analysis. Stroke. 2003;34:1710–6.
10. Zhang H, et al. Prevalence of superficial Siderosis in patients with cerebral amyloid Angiopathy.
Neurology. 2010;74:1346–50.
11. Charidimou A, et al. Cortical superficial siderosis and intracerebral hemorrhage risk in cerebral
amyloid angiopathy. Neurology. 2013;81:1666–73.
12. Wilson D, et al. Recurrent stroke risk and cerebral microbleed burden in ischemic stroke and
TIA. Neurology. 2016;87(14):1501–10.
13. Poli D, et al. Recurrence of ICH after resumption of anticoagulation with VK antagonists:
CHIRONE Study. Neurology. 2014;82:1020–6.
14. Gorelick PB, Weisman SM. Risk of hemorrhagic stroke with aspirin use: an update. Stroke.
2005;36:1801–7.
227
5. Viswanathan A, et al. Antiplatelet use after intracerebral hemorrhage. Neurology. 2006;66:206–9.
1
16. Ruff CT, et al. Comparison of the efficacy and safety of new oral anticoagulants with warfarin in
patients with atrial fibrillation: a meta-analysis of randomised trials. Lancet. 2014;383:955–62.
17. Hart RG, et al. Intracranial hemorrhage in atrial fibrillation patients during anticoagulation with
warfarin or dabigatran: the RE-LY trial. Stroke. 2012;43:1511–7.
18. Majeed A, Kim Y-K, Roberts RS, Holmström M, Schulman S. Optimal timing of resumption of
warfarin after intracranial hemorrhage. Stroke. 2010;41:2860–6.
228
SUBARACHNOID HEMORRHAGE – DIFFERENTIAL
Catherine S. W. Albin and Sahar F. Zafar
While aneurysm rupture is the classic and most feared etiology of subarachnoid
hemorrhage (SAH), trauma is the most common. However, there are many other
additional etiologies of SAH. In addition to evaluating for aneurysm and trauma,
the following etiologies should also be considered:
□□ Coagulopathy – can result in spontaneous SAH.
□□ Reversible cerebral vasoconstriction syndrome (RCVS) – commonly results in
convexity SAH, see page 123.
□□ Venous sinus thrombosis – may be associated with both SAH and parenchy-
mal hemorrhage.
□□ Endocarditis – may be the result of mycotic aneurysm rupture or hemorrhagic
ischemia from septic emboli.
□□ Intracranial dissection – See page 89.
□□ Cerebral amyloid angiopathy (CAA) – mechanism likely overlaps with process
causing superficial siderosis.
□□ Pituitary apoplexy.
□□ Bleeding from other vascular malformations – including arteriovenous malfor-
mations and dural arteriovenous fistulas.
Perimesencephalic Non-Aneurysm SAH Subarachnoid blood in the interpeducu-
lar, crural, ambient, and quadrigeminal cisterns, typically located immediately anterior
to the midbrain or in the prepontine cistern. These patients tend to be younger and
less hypertensive than patients with aneurysmal hemorrhage. They have a more
benign clinical course than angio-confirmed aneurysmal SAH with a subsequent
rebleed rate of 2–5%, as well as lower rates of hydrocephalus and of significant
vasospasm. However, they may still develop significant hyponatremia and cardiac
abnormalities [1].
Perimesencephalic blood in the prepontine and Convexity SAH, in this case from reversible
intrapeduncular cisterns. cerebral vasoconstriction syndrome.
REFERENCE
1. Kapadia A, et al. Nonaneurysmal perimesencephalic subarachnoid hemorrhage: diagno-
sis, pathophysiology, clinical characteristics, and long-term outcome. World Neurosurg.
2014;82(6):1131–43.
230
ANEURYSMAL SAH – ADMISSION AND EARLY
MANAGEMENT
Christopher Reeves and Catherine S. W. Albin
The care of aneurysmal SAH is generally a two-staged approach. The first stage
occurs when the patient is “unsecured” meaning there has been no neurosurgical
intervention on the aneurysm. The objective of this stage is to prevent herniation
from hydrocephalus and cerebral edema and then to identify and intervene upon
the source of bleeding. It is critical to prevent re-rupture of the suspected aneu-
rysm; outcomes following rebleed events are markedly worse.
REFERENCES
1. Connolly J, Sander E, et al. Guidelines for the management of aneurysmal subarachnoid hemor-
rhage: a guideline for healthcare professionals from the American Heart Association/American
Stroke Association. Stroke. 2012;43(6):1711–37.
2. Hillman J, et al. Immediate administration of tranexamic acid and reduced incidence of early
rebleeding after aneurysmal subarachnoid hemorrhage: a prospective randomized study. J
Neurosurg. 2002;97(4):771–8.
3. Diringer MN, Bleck TP, Claude Hemphill J 3rd, et al. Critical care management of patients fol-
lowing aneurysmal subarachnoid hemorrhage: recommendations from the Neurocritical Care
Society’s Multidisciplinary Consensus Conference. Neurocrit Care. 2011;15(2):211–40.
232
SUBARACHNOID HEMORRHAGE – SCORING SYSTEMS
Catherine S. W. Albin and Sahar F. Zafar
234
ANEURYSMAL SAH – DAILY MANAGEMENT PRINCIPLES
Christopher Reeves and Catherine S. W. Albin
CT angiography axial MIPs day 1. Distal CT angiography axial MIPS day 9. Flow-diverting stent
ICA aneurysm is one CT slice below. noted with arrow. Significant vasospasm throughout the
Image demonstrates no vasospasm or anterior and posterior circulations, but most prominent
baseline intracranial atherosclerosis in the bilateral MCAs in this projection (arrow heads).
236
• Close neurologic monitoring with very frequent neuro-checks.
• Institutions vary in what DCI monitoring is performed which may include some
combination of CT angiography (CTA), CT perfusion (CTP), brain tissue oxygen
monitoring, or Near Infared Spectroscopy (NIRS).
• Many institutions rely on daily or near daily transcranial dopplers (TCDs) as a key
component of vasospasm detection (see
page 47 for interpretation).
• Consider cvEEG for Hunt Hess 3–5 patients Percent Alpha Variability:
as a component of DCI monitoring or where A qualitative analysis of the
there is suspicion for non-convulsive seizures. change in Alpha variability. 4
○ Trend the relative alpha variability (rAV) is excellent, 3 is good, 2 is
and alpha–delta ratio (ADR), worsening fair, and 1 is poor. Any
focal slowing, and new epileptiform abnor‑ decrease in the score
malities. Late appearing epileptiform should heighten concern
abnormalities had the highest predictive for DCI.
valve for DCI [2]. Alpha–Delta Ratio:
○ Changes in TCDs and EEG may not cor‑ Concerning when there is a
relate with exam changes, but should decrement in the alpha
heighten vigilance about impending DCI. percentage (i.e. more
slowing) especially when
the difference is lateralized.
TREATMENT AND PREVENTION OF DELAYED
CEREBRAL ISCHEMIA
• At the time of admission, begin nimodipine
60 mg q4h for 21 days.
○ May change to 30 mg q2h if larger dose results in hypotension.
• If clinical decline, urgently communicate with neurosurgery and initiate rescue
therapies:
○ Position patient flat or reverse Trendelenburg for brain perfusion as respiratory
status allows.
○ Ensure euvolemia – bolus as needed. (Hypervolemia – part of the now aban‑
doned “triple-H therapy” – has had questionable benefit and may cause cardio‑
pulmonary complications) [7].
○ Consider induced hypertension with vasopressors.
○ CT/CTA/CTP may be pursued to evaluate for vasospasm and infarction prior to
DSA; if there is a high concern, however, the patient may go directly to DSA for
intra-arterial therapies. Notify neuroendovascular/neurosurgery immediately.
• Intra-arterial therapies include IA-milrinone or IA-verapamil and angioplasty.
237
• There are no large, randomized trials that address the management of vasospasm
discovered by TCDs or CT angiography with only a questionable clinical exam
correlate. However, emerging therapies and protocols have suggested some
benefit in the prevention and treatment of vasospasm, and may be considered
prior to or in addition to intra-arterial therapy.
CEREBRAL EDEMA
• May be seen early or late in the course, with 8–67% of patients demonstrating
early global cerebral edema and 12% of patients developing edema in a delayed
fashion [8].
• Mechanisms include early ischemic injury at the time of aneurysm rupture, dysfunc‑
tion of autoregulation, reaction to toxic degrading blood products, neuroinflamma‑
tion, and hyponatremia [9].
• Management should include ICP monitoring and treatment of cerebral edema in a
stepwise fashion, see page 187, hypertonic saline is the preferred hyperosmolar
agent in SAH.
SYSTEMIC COMPLICATION
Fever
• Extremely common in aSAH patients, associated with poor outcomes, but unclear
if treatment improves outcomes.
• Infectious and DVT screening should occur routinely in all febrile patients, but also
consider drug withdrawal, neuroleptic malignant syndrome, and serotonin syn‑
drome in appropriate patients.
• Treat with acetaminophen, bromocriptine, and, if needed, consider surface cooling
or intravascular devices.
• Treat shivering which can raise ICP.
238
• In general, AVOID free water restrictions, as the goal is euvolemia, given the risk of
vasospasm/DCI.
• With SIADH use salt tabs or 3% sodium chloride infusion.
• With CSW can use salt tabs, 3% sodium chloride infusion, and/or mineralocorti‑
coids if needed to ensure euvolemia.
Other Complications
Hyperglycemia, pulmonary edema, cardiomyopathy and EKG changes, hypotha‑
lamic, pituitary disruption, and anemia are all common in subarachnoid hemorrhage
patients and need to be monitored for. The specific management is beyond the
scope of this chapter.
REFERENCES
1. Chung DY, Mayer SA, Rordorf GA. External ventricular drains after subarachnoid hemorrhage: is
less more? Neurocrit Care. 2018;28(2):157–61.
2. Rosenthal ES, et al. Continuous electroencephalography predicts delayed cerebral ischemia
after subarachnoid hemorrhage: a prospective study of diagnostic accuracy. Ann Neurol.
2018;83(5):958–69.
3. Lannes M, et al. Milrinone and homeostasis to treat cerebral vasospasm associated with
subarachnoid hemorrhage: the Montreal Neurological Hospital protocol. Neurocrit Care.
2012;16(3):354–62.
4. Webb A, Kolenda J, Martin K, Wright W, Samuels O. The effect of intraventricular administra‑
tion of nicardipine on mean cerebral blood flow velocity measured by transcranial Doppler in
the treatment of vasospasm following aneurysmal subarachnoid hemorrhage. Neurocrit Care.
2010;12(2):159–64.
5. Suzuki M, Mamoru D, Yasunari O, Ogasawara K, Ogawa A. Intrathecal administration of
nicardipine hydrochloride to prevent vasospasm in patients with subarachnoid hemorrhage.
Neurosurg Rev. 2001;24:180–4.
6. Senbokya, N et al. “Effects of cilostazol on cerebral vasospasm after aneurysm subarachnoid
hemorrhage: a multicenter prospective, randomized, open-label blinded end point trial.”
J Neurosurg. 2013;118(1):121–30.
7. Rinkel GJ, Feigin VL, Algra A, van Gijn J. Circulatory volume expansion therapy for aneurysmal
subarachnoid haemorrhage. Cochrane Database Syst Rev. 2004;2004:CD000483.
8. Claassen J, Carhuapoma JR, Kreiter KT, Du EY, Connolly ES, Mayer SA. Global cerebral
edema after subarachnoid hemorrhage: frequency, predictors, and impact on outcome. Stroke.
2002;33:1225–32.
9. Hayman EG, et al. Mechanisms of global cerebral edema formation in aneurysmal subarachnoid
hemorrhage. Neurocrit Care. 2017;26(2):301–10.
239
SUBARACHNOID HEMORRHAGE – NOTABLE TRIALS
Catherine S. W. Albin and Sahar F. Zafar
242
THEME TRIAL NAME TRIAL DESIGN MAJOR FINDINGS
Magnesium MASH-2 Trial [8] Phase 3. Internal, 158 patients (26.2%) in the
(Lancet 2012) multicenter, randomized. magnesium group had a
1204 patients randomized poor outcome versus 151
to same treatment, but (25.3%) in the placebo
primary outcome was poor group. Nonsignificant
outcome (mRS 4–6) difference by treatment
3 months after SAH. category.
Intravenous The Montreal Protocol Large case series. Milrinone Large case series, thus no
Milrinone (Neurocritical Care was used as part of a control. However, no
2012) [9] larger protocol focused on significant side effects and
maintaining fluid and the infusion duration was on
electrolyte homeostasis. average 9.8 days; 68% of
Patients who developed patients required
new neurologic deficits norepinephrine. 48.9% of
were treated with patients were able to return
crystalloids to a CVP ≧ 6, to previous activities and
electrolytes were corrected 75% of patients had a good
and a milrinone 0.1–0.2 outcome (mRS ≦ 2).
mg/kg bolus was given; an
infusion was then initiated
at 0.75 mcg/kg/min and
increased if tolerated to 1
mcg/kg/hour.
Dedicated Impact of a dedicated 703 patients with aSAH Patients treated by a
NeuroICU neurocritical care team retrospectively reviewed for neurocritical care team
care in treating patients discharge outcomes before were significantly more
with aSAH [10] and after the development likely to be discharged
(Neurocrit Care 2012) of a multidisciplinary home (36.5% vs. 25.2%)
neurocritical care team. and were more likely to
receive definitive aneurysm
treatment.
REFERENCES
1. Molyneux A, Kerr R, Stratton I, et al. International Subarachnoid Aneurysm Trial (ISAT) of neu-
rosurgical clipping versus endovascular coiling in 2143 patients with ruptured intracranial aneu-
rysms: a randomised trial. Lancet. 2002;360:1267–74.
2. Hillman J, Fridriksson S, Nilsson O, Yu Z, Saveland H, Jakobsson KE. Immediate administration
of tranexamic acid and reduced incidence of early rebleeding after aneurysmal subarachnoid
hemorrhage: a prospective randomized study. J Neurosurg. 2002;97:771–8.
3. Post R, et al. Ultra-early tranexamic acid after subarachnoid haemorrhage (ULTRA): a ran-
domised controlled trial. Lancet. 2020;397(10269):112–8.
243
4. Allen GS, et al. Cerebral arterial spasm–a controlled trial of nimodipine in patients with subarach-
noid hemorrhage. N Engl J Med. 1983;308(11):619–24.
5. Dorhout Mees SM, Rinkel GJ, Feigin VL, et al. Calcium antagonists for aneurysmal subarachnoid
haemorrhage. Cochrane Database Syst Rev. 2007;3:CD000277.
6. Treggiari MM, et al. Systematic review of the prevention of delayed ischemic neurological deficits
with hypertension, hypervolemia, and hemodilution therapy following subarachnoid hemorrhage.
J Neurosurg. 2003;98(5):978–84.
7. Kirkpatrick PJ, et al. Simvastatin in aneurysmal subarachnoid haemorrhage (STASH): a multicen-
tre randomised phase 3 trial. Lancet Neurol. 2014;13(7):666–75.
8. Mees SMD, Algra A, Vandertop WP, et al. Magnesium for aneurysmal subarachnoid haemor-
rhage (MASH-2): a randomised placebo-controlled trial. Lancet. 2012;380(9836):44–9.
9. Lannes M, et al. Milrinone and homeostasis to treat cerebral vasospasm associated with
subarachnoid hemorrhage: the Montreal Neurological Hospital protocol. Neurocrit Care.
2012;16(3):354–62.
10. Samuels O, Webb A, Culler S, Martin K, Barrow D. Impact of a dedicated neurocritical care team
in treating patients with aneurysmal subarachnoid hemorrhage. Neurocrit Care. 2011;14:334–40.
244
TRAUMATIC BRAIN INJURY
Catherine S. W. Albin and Sahar F. Zafar
EMERGENT MANAGEMENT
246
PRINCIPLES OF ICU MANAGEMENT
247
SURGICAL MANAGEMENT IN TBI
Traumatic brain injury patients should be co-managed with neurosurgeons.
Focal Lesions
SUBDURAL HEMATOMA [4] EPIDURAL HEMATOMA
Guidance for evacuation − Epidural blood >30 cc
− SDH >1 cm or midline shift >5 mm − Acute epidural hematoma poor neurologic
− GCS ≦ 8 exam or anisocoria
− GCS decreased by two or more points
between injury and hospital admission
− Asymmetric or fixed pupils
248
It should be sought in patients with chronic subdural hematomas. A meta-analysis
demonstrated that hematoma recurrence rate was significantly lower in the emboliza-
tion group (2.1% vs 27.7%) [7].
REFERENCES
1. Carney N, et al. Guidelines for the management of severe traumatic brain injury. Neurosurgery.
2017;80(1):6–15.
2. Al-Mufti F, Mayer SA. Neurocritical care of acute subdural hemorrhage. Neurosurg Clin.
2017;28(2):267–78.
3. Haghbayan H, et al. The prognostic value of MRI in moderate and severe traumatic brain injury:
a systematic review and meta-analysis. Crit Care Med. 2017;45(12):e1280–8.
4. Bullock MR, et al. Surgical management of acute subdural hematomas. Neurosurgery.
2006;58(suppl_3):S2–16.
5. Hutchinson PJ, et al. Trial of dexamethasone for chronic subdural hematoma. N Engl J Med.
2020;383(27):2616–27.
6. Ban SP, et al. Middle meningeal artery embolization for chronic subdural hematoma. Radiology.
2018;286(3):992–9.
7. Srivatsan A, et al. Middle meningeal artery embolization for chronic subdural hematoma: meta-
analysis and systematic review. World Neurosurg. 2019;122:613–9.
249
TRIALS IN TBI
Catherine S. W. Albin and Sahar F. Zafar
REFERENCES
1. Cooper DJ, Rosenfeld JV, Murray L, et al. Decompressive craniectomy in diffuse traumatic brain
injury. N Engl J Med. 2011;364:1493–502.
2. Hutchinson PJ, et al. Trial of decompressive Craniectomy for traumatic intracranial hypertension.
N Engl J Med. 2016;375(12):1119–30.
3. CRASH Trial Collaborators. Effect of intravenous corticosteroids on death within 14 days in
10 008 adults with clinically significant head injury (MRC CRASH trial): randomised placebo-
controlled trial. Lancet. 2004;364(9442):1321–8.
4. Chesnut RM, et al. A trial of intracranial-pressure monitoring in traumatic brain injury. N Engl J
Med. 2012;367(26):2471–81.
5. CRASH, The. Effects of tranexamic acid on death, disability, vascular occlusive events and other
morbidities in patients with acute traumatic brain injury (CRASH-3): a randomised, placebo-
controlled trial. Lancet. 2019;394(10210):1713–23.
252
NEUROPROGNOSIS AND INDUCED NORMOTHERMIA
AFTER CARDIAC ARREST
Priya Srikanth and Catherine S. W. Albin
The optimal degree of temperature control after cardiac arrest is still unknown. Prior
evidence from small studies had suggested that for out of hospital VT/VF arrest that
33°C was superior to no TTM [1, 2]. In 2013, TTM1 (Targeted temperature
management at 33°C versus 36°C after cardiac arrest [3]) compared 950 patients
with out of hospital arrest and found that 36°C was equivalent to 33°C. Thus, there
was evidence to support a less severe degree of hypothermia.
However, in 2021 the TTM2 Trial (Target hypothermia versus targeted normothermia
after out-of-hospital cardiac arrest, NEJM) [4] was published which compared 2000
patients randomized to 36°C to fever control (the patients only were temperature-
managed if they developed a temperature >37.8°C and the target was set to 37.5°C).
In this trial, neuroprognostication was delayed for 96 hours and the clinicians
performing the neuroprognostication were blinded. Unexpectedly, there was no
mortality benefit of 36°C and there was no difference in disability at 6 months and
there was a signal of harm for hypothermia (more arrhythmias and longer ventilation
times). The lack of benefit in hypothermia held true across subgroup analysis.
At this point, literature supports aggressive fever prevention in all brain injured
patients and there may be a subset of patients for whom hypothermia could have
benefit. Some clinicians may still opt to perform mild hypothermia to 36°C and fever
prevention remains critically important, pending TTM3.
254
HISTORY IMPORTANT IN NEUROPROGNOSIS:
□□ Time of arrest
□□ Time from arrest to CPR
□□ Initial recorded rhythm
□□ Minutes without a pulse
□□ Time of ROSC
□□ Suspected etiology
□□ Medications given during arrest
PERSPECTIVE IN NEUROPROGNOSTICATION
Current guidelines recommend against withdrawal of life-sustaining because
of perceived poor neurologic prognosis (WLST-N) before 72 hours, and neuro-
prognostication should not be done until after that time.
However, a multicenter trial of OHCA subjects found that 1/3 of patients who died in
the hospital died because of WLST-N before 72 hours. By propensity matching and
the use of logistic regression models, this study estimated that 16% of patients in
whom life support was withdrawn may have had a functionally favorable survival [5].
It is critically important to take time and collect all possible variables for this
matter of life and death.
255
PREDICTOR FPR FOR POOR PROGNOSIS IN TTM
Non-VF cardiac arrest 15% (6–30%)
ROSC >25 mins 24% (13–40%)
256
REFERENCES
1. Hypothermia after Cardiac Arrest Study Group. Mild therapeutic hypothermia to improve the neu-
rologic outcome after cardiac arrest. NEJM. 2002;346(8):549–56.
2. Bernard SA, et al. Treatment of comatose survivors of out-of-hospital cardiac arrest with induced
hypothermia. NEJM. 2002;346(8):557–63.
3. Nielsen N, et al. Targeted temperature management at 33°C versus 36°C after cardiac arrest.
NEJM. 2013;369(23):2197–206.
4. Dankiewicz J, et al. Hypothermia versus normothermia after out-of-hospital cardiac arrest. N Engl
J Med. 2021;384(24):2283–94.
5. Elmer J, et al. Association of early withdrawal of life-sustaining therapy for perceived neurological
prognosis with mortality after cardiac arrest. Resuscitation. 2016;102:127–35.
6. Zhou SE, et al. Distinct predictive values of current Neuroprognostic guidelines in post- cardiac
arrest patients. Resuscitation. 2019;139:343–50.
7. Wu O, et al. Predicting clinical outcome in comatose cardiac arrest patients using early noncon-
trast computed tomography. Stroke. 2011;42(4):985–92.
8. Oddo M, Sandroni C, Citerio G, et al. Quantitative versus standard pupillary light reflex for early
prognostication in comatose cardiac arrest patients: an international prospective multicenter
double-blinded study. Intensive Care Med. 2018;44(12):2102–11.
9. Elmer J, Rittenberger JC, Faro J, Molyneaux BJ, Popescu A, Callaway CW, Baldwin M, Pittsburgh
Post-Cardiac Arrest Service. Clinically distinct electroencephalographic phenotypes of early
myoclonus after cardiac arrest. Ann Neurol. 2016;80:175–84.
10. Westover MB, Edlow BL, Greer DM. Coma after cardiac arrest: management and neurological
prognostication. London: MGH Cardiology Board Review. Springer; 2014. p. 471–85.
11. Tiainen M, Kovala TT, Takkunen OS, Roine RO. Somatosensory and brainstem auditory evoked
potentials in cardiac arrest patients treated with hypothermia. Crit Care Med. 2005;33(8):1736–40.
12. Hirsch KG, et al. Prognostic value of diffusion-weighted MRI for post-cardiac arrest coma.
Neurology. 2020;94(16):e1684–92.
13. Stammet P, Collignon O, Hassager C, Wise MP, Hovdenes J, Åneman A, et al. Neuron-specific
enolase as a predictor of death or poor neurological outcome after out-of-hospital cardiac arrest
and targeted temperature management at 33 degrees C and 36 degrees C. J Am Coll Cardiol.
2015;65(19):2104–14.
14. Gillick K, Rooney K. Serial NSE measurement identifies non-survivors following out of hospital
cardiac arrest. Resuscitation. 2018;128:24–30.
257
STATUS EPILEPTICUS
Catherine S. W. Albin and Sahar F. Zafar
DEFINITIONS
Convulsive status epilepticus: >5 mins of convulsive seizures or ≧2 seizures
without return to baseline.
Nonconvulsive status epilepticus (NCSE): Multiple definitions exist using electro-
graphic and electroclinical data. Generally, NCSE is defined as rhythmic/periodic
EEG activity with evolution and with clear correlation between the EEG and clinical
symptoms. A benzodiazepine trial may be helpful to determine the correlation
between clinical and EEG findings (see page 263 for more information on the
interictal continuum and nonconvulsive status epilepticus).
Refractory status epilepticus (RSE): Status that continues despite stages I and II
treatment (see below).
Superrefractory status epilepticus (SRSE): Status that continues despite treat-
ment with anesthetics for >24 hours.
For all treatment groups, the success rate – the absence of clinically apparent
seizures with improving responsiveness at 60 mins after the start of the infusion –
was <50%
*In the TRENDS trial [3], lacosamide was non-inferior to fosphenytoin in controlling
nonconvulsive seizures. Given the favorable side-effect profile and low drug-drug
interaction, it has gained popularity but was not part of ESETT
Practical pearls for the dosing, dose-adjustment, and commonly
encountered side effects for levetiracetam, valproate, and
fosphenytoin can be found on page 141
− For VPA, PHT, and PHB, check level 1 hour after the load (free PHT level is
preferable, but send total if that will result more quickly)
− For fos-PHT send level 2 hours after load (free PHT level is preferable, but
send total if that will result more quickly)
260
Phase III: Anti-seizure doses of anesthetics will be higher than what is needed for general
sedation
For IV anesthetics, bolus at the initiation of treatment and consider re-bolusing for
breakthrough seizures before increasing the maintenance dose
Fourth-line Ketamine: Gaspard et al. [4] in a multicenter study found permanent control of
treatments: RSE in 57% of episodes; ketamine appeared to have contributed to the control in
32% of patients. Alkhachroum et al. [5] found that seizure burden decreased by
50% within 24 hours of ketamine infusion in 81% (55 of 68) of patients
retrospectively studied. It may cause hypertension and tachycardia and also
cardiac depression at high doses
REFERENCES
1. Glauser T, et al. Evidence-based guideline: treatment of convulsive status epilepticus in children
and adults: report of the guideline Committee of the American Epilepsy Society. Epilepsy Curr.
2016;16(1):48–61.
2. Chamberlain JM, et al. Efficacy of levetiracetam, fosphenytoin, and valproate for established
status epilepticus by age group (ESETT): a double-blind, responsive-adaptive, randomised con-
trolled trial. Lancet. 2020;395(10231):1217–24.
3. Husain AM, et al. Randomized trial of lacosamide versus fosphenytoin for nonconvulsive sei-
zures. Ann Neurol. 2018;83(6):1174–85.
4. Gaspard N, et al. Intravenous ketamine for the treatment of refractory status epilepticus: a retro-
spective multicenter study. Epilepsia. 2013;54(8):1498–503.
5. Alkhachroum A, et al. Ketamine to treat super-refractory status epilepticus. Neurology.
2020;95(16):e2286–94.
6. Gaspard N, et al. New-onset refractory status epilepticus: etiology, clinical features, and outcome.
Neurology. 2015;85(18):1604–13.
7. Thakur KT, et al. Ketogenic diet for adults in super-refractory status epilepticus. Neurology.
2014;82(8):665–70.
8. Niedermeyer E, Freund G, Krumholz A. Subacute encephalopathy with seizures in alcoholics: a
clinical-electroencephalographic study. Clin Electroencephalogr. 1981;12(3):113–29.
9. Williams TJ, et al. Association of autoimmune encephalitis with combined immune checkpoint
inhibitor treatment for metastatic cancer. JAMA Neurol. 2016;73(8):928–33.
10. Akbik F, et al. The PET sandwich: using serial FDG-PET scans with interval burst suppression to
assess ictal components of disease. Neurocrit Care. 2020;33(3):657–69.
262
CONTINUOUS EEG MONITORING, ELECTROGRAPHIC
SEIZURES, AND THE ICTAL-INTERICTAL CONTINUUM
Catherine S. W. Albin and Sahar F. Zafar
Continuous EEG is also used in vasospasm monitoring – see page 237 for more details.
TERMS
The American Clinical Neurophysiology Society recently updated their standardiza-
tion for critical care EEG terminology [1]. The wide variety of features that may be
reported in a continuous EEG report is outside the scope of this chapter. However,
below is a summary of what will be included in a continuous EEG report. For more
information and further details, see the ACNS terminology guidelines and refer to the
Salzburg criteria for seizures [2].
264
EXAMPLES OF COMMONLY ENCOUNTERED CONTINUOUS EEG FINDINGS
LATERALIZED PERIODIC DISCHARGES (LPDS) LATERALIZED RHYTHMIC DELTA ACTIVITY (LRDA)
Lateralized sharp waves or spikes made have Usually reflects the presence of a focal lesion;
associated slow waves. Commonly encountered in associated with the risk of acute seizures,
stroke, intracerebral hemorrhage, subarachnoid especially nonconvulsive status epilepticus
hemorrhage, tumors, abscesses, Creutzfeldt-Jakob
disease, herpes simplex virus, and other
infectious/autoimmune pathology. LPDs are highly
associated with seizures especially in the setting of
acute illness, metabolic disturbances, or focal
lesions
265
2HELPS2B SCORE [3]
Predicts seizure risk. The authors propose the 2HELPS2B score can be reported
after 1 hour of screening with IV sedation minimized.
Score = 0, cEEG not needed (although 90 mins of screening should be considered in
patients with coma).
Score = 1, at least 12 hours of monitoring. If the score increases to ≥2 during
12 hours, monitor at least 24 hours.
Score ≥ 2, at least 24 hours of cEEG.
RISK FACTOR SCORE
Frequency >2hza 1 Predicted Seizure Riskd
Independent sporadic epileptiform discharges 1 0 = <5%
LPD/BIPD/LRDA 1 1 = 12
Plus features (superimposed rhythmic, fast, 1 2 = 27%
sharp)b 3 = 50%
Prior seizurec 1 4 = 73%
Bilateral independent periodic discharges 2
5 + = 88%
Total score 0–7
a
Frequency of any periodic or rhythmic pattern of more than
2 Hz except generalized rhythmic delta activity
b
Plus features include superimposed rhythmic, fast, or sharp
activity only on LRDA, LPDs, or BIPDs
c
Prior seizure includes a remote history of epilepsy or recent
events suspicious for clinical seizures
d
Predicted seizure risk based on the 2HELPS2B model
*If the EEG improves with administration of AED but there is no clinical improvement,
then continue to monitor as clinical improvement may be delayed. Continued trial of
AED is likely warranted.
**If the EEG continues to have features of the IIC, it is important to use clinical
judgment. Can consider a longer AED trial, advanced neuroimaging as with SPECT
or PET, or discontinue AEDs and continue to evaluate for an underlying cause such
as a toxic-metabolic, infection, and structural abnormality that may contribute to or
cause “cortical irritability.”
REFERENCES
1. Hirsch LJ, et al. American Clinical Neurophysiology Society’s Standardized Critical Care EEG
Terminology: 2021 Version. J Clin Neurophysiol. 2021;38(1):1–29.
2. Leitinger M, Trinka E, Gardella E, et al. Diagnostic accuracy of the Salzburg EEG criteria for non-
convulsive status epilepticus: a retrospective study. Lancet Neurol. 2016;15:1054–62.
3. Struck AF, Ustun B, Ruiz AR, et al. Association of an electroencephalography-based risk score
with seizure probability in hospitalized patients. JAMA Neurol. 2017;74(12):1419–24.
4. Rodríguez V, Rodden MF, LaRoche SM. “Ictal–interictal continuum: a proposed treatment algo-
rithm.” Clin Neurophysiol. 2016;127(4):2056–64.
267
NEUROMUSCULAR CRISES: ICU MANAGEMENT
OF GUILLAIN-BARRÉ SYNDROME AND MYASTHENIA
GRAVIS
Catherine S. W. Albin and Sahar F. Zafar
GUILLAIN-BARRÉ SYNDROME
Polyneuropathy is characterized classically as ascending weakness usually following
a respiratory or GI illness with areflexia and high protein in the CSF without elevation
in cell count.
Respiratory failure occurs in 20–30% of patients with Guillain-Barré syndrome [2].
The need for intubation should be highly considered if [3]:
–– The forced vital capacity (FVC) is <20 ml/kg (~1.5 L in a 70 kg person). A very
rough estimate is having the patient count as high as they can in 1 breath.
Counting to 10 = 1 L, counting to 20 = 2 L.
–– Negative inspiratory force (NIF) of > –30 cm H2O (meaning less negative; 20 −
would be a concerning number).
–– Reduction of these numbers by >30% in a brief time.
–– It is also important to assess neck flexion and extension as these can be proxies
for diaphragmatic weakness.
Note that ABGs and pulse oximetry readings are not sensitive for impending
respiratory failure. Intervention should be persued before the patient is hypercarbic
or hypoxic. Noninvasive ventilation is not appropriate in most cases as it cannot be
used for the duration of time the patients need for recovery. Intubation is likely to be
prolonged in Guillain-Barré syndrome, and in many cases early tracheostomy should
be considered.
Dysautonomia complicates up to 70% of patients in the ICU and may be manifested
by arrhythmias, diaphoresis, labile blood pressure, gastroparesis, urinary retention,
and ileus.
IVIG PLEX
Pros More easily administered (no central line Small study in children demonstrating that in
required) mechanically ventilated patients, PLEX resulted
in shorter duration of ventilation and a
tendency toward shorter PICU stay [5]
Cons Side effects include headache, aseptic Requires a pheresis catheter
meningitis, hypercoagulability resulting in
Citrate-induced hypocalcemia: check iCal
thromboembolic complications, infusion site
during procedure, cardiac monitoring;
reaction, and fever. Anaphylaxis is
citrate-induced metabolic alkalosis
extremely rare; urticaria, flushing, pain, and
nausea/vomiting are normally rate related. Removal of highly protein-bound drugs.
TRALI and TACO are both possible but rare
Allergic reaction to administered FFP if being
Important that patients are well hydrated used; TRALI
before the infusion to prevent thrombosis
Non-plasma replacement fluids may cause
and renal complications
hypokalemia, hypofibrinogenemia and
coagulation factor depletion.
MYASTHENIA GRAVIS
Pathology occurs via an immune attack against nicotinic acetylcholine receptors at
the neuromuscular junction or related proteins (MUSK = muscle-specific kinase;
LRP4 = low-density lipoprotein receptor-related protein). This causes a fatigable
weakness which frequently involves the bulbar neuromuscular junction resulting in
oropharyngeal weakness (high aspiration risk) and diaphragmatic weakness.
270
Workup
□□In patients without a prior diagnosis or without prior screening, CT chest
imaging should be pursued to rule out the presence of a thymoma.
□□ In patients on treatment, differentiate between a myasthenic crisis and cholin-
ergic crisis from overmedication with cholinesterase inhibitors such as pyr-
idostigmine (Mestinon). Clues to overtreatment with pyridostigmine include
diarrhea, nausea, urinary incontinence, blurred vision and abdominal cramps in
addition to muscle weakness.
ICU Management
Intubation may be needed because of diaphragmatic weakness or because of
difficulty managing secretions/aspiration risk. Patients need close monitoring of neck
flexion/extension weakness, FVC/NIFs (although bulbar weakness may impair the
ability to generate a good seal), cough, and swallowing.
–– Noninvasive ventilation can be considered in patients with mild secretions to
help alleviate work of breathing and reduce ventilator days [6]; intubation should
be pursued if patient has copious oral secretions, weak cough, and if there is
hypercarbia/hypoxia.
–– Intubation should be pursued if the patient’s vital capacity (VC) falls below 15 mL/
kg [7] or for any concern for aspiration or stridor.
–– Do not use succinylcholine for intubation.
–– Stop pyridostigmine when the patient is intubated (to decrease bronchial sec-
tions), but it can be restarted when the patient is ready for ventilator weaning,
usually started at half of the home dose and uptitrated.
–– Patients with new-onset MG should be evaluated for thymoma, although the tim-
ing of thymectomy is debated as the benefit is not immediate.
Treatment
PLEX or IVIG should be initiated urgently in patients suspected of myasthenic crisis.
Plasma exchange is generally preferred as it produces a rapid improvement in 75%
of patients [8]. Steroids may worsen weakness acutely and thus are usually avoided
in crisis but may be considered if the patient is intubated and started in conjunction
with or after initiation of PLEX or IVIG [9]. Prednisone 1 mg/kg/day is the usual
starting dose and is often continued to 4 weeks after the exacerbation. Starting
treatment with a long-term immunosuppressant such as azathioprine, mycopheno-
late mofetil, or cyclosporine should be discussed with the provider that will follow the
patient longitudinally. Newer immune therapies like eculizumab or rituximab may be
pursued for maintenance immunosuppression although this is not typically done
during the ICU phase of illness.
For a list of medications that should be avoided in patients with myasthenia,
see page 343.
271
REFERENCES
1. Damian MS, Wijdicks EFM. The clinical management of neuromuscular disorders in intensive
care. Neuromuscul Disord. 2019;29(2):85–96.
2. Damian MS, et al. The effect of secular trends and specialist neurocritical care on mortality for
patients with intracerebral haemorrhage, myasthenia gravis and Guillain–Barré syndrome admit-
ted to critical care. Intensive Care Med. 2013;39(8):1405–12.
3. Lawn ND, Fletcher DD, Henderson RD, Wolter TD, Wijdicks EF. Anticipating mechanical ventila-
tion in Guillain–Barré syndrome. Arch Neurol. 2001;58(6):893–8.
4. Ortiz-Salas P, et al. Human immunoglobulin versus plasmapheresis in Guillain–Barre syndrome
and myasthenia gravis: a meta-analysis. J Clin Neuromuscul Dis. 2016;18(1):1–11.
5. El-Bayoumi MA, et al. Comparison of intravenous immunoglobulin and plasma exchange in treat-
ment of mechanically ventilated children with Guillain Barré syndrome: a randomized study. Crit
Care. 2011;15(4):1–6.
6. Seneviratne J, et al. Noninvasive ventilation in myasthenic crisis. Arch Neurol. 2008;65(1):54–8.
7. Fink ME. Treatment of the critically ill patient with myasthenia gravis. In: Ropper AH, editor.
Neurological and neurosurgical intensive care. 3rd ed. New York: Raven Press; 1993. p. 351–62.
8. Mayer S. Intensive care of the myasthenic patient. Neurology. 1997;48(Suppl 5):70S–5S.
9. Green DM. Weakness in the ICU: Guillain–Barré syndrome, myasthenia gravis, and critical ill-
ness polyneuropathy/myopathy. Neurologist. 2005;11(6):338–47.
272
EVALUATION OF C-SPINE TRAUMA
Catherine S. W. Albin and Sahar F. Zafar
C-SPINE INJURIES
–– Mechanisms of injury include flexion, flexion-rotation, extension, and vertical
compression.
–– Any patient with a neurologic deficit or radiographic evidence of injury should be
presumed to have an unstable fracture until further workup is completed.
Fig. 52.1 Sagittal CT scan on bone window demonstrating a Dens Fracture: Type 2
274
CANADIAN RULES FOR C-SPINE CLEARANCE IN THE ADULT PATIENT [1]
Note the patient must have a GCS of 15 and be hemodynamically stable.
275
C-SPINE CLEARANCE IN THE OBTUNDED TRAUMA PATIENT
Trauma patients with depressed neurologic status should have CT C-spine imaging
performed. Removal of the collar in this scenario is much more complicated as the
patient cannot endorse pain. There is no consensus about best practice in this
scenario. If the neurologic deficit is expected to improve (such as intoxication), wait
until the patient can be more fully assessed.
REFERENCES
1. Steill IG, Wells GA, Vademheen KL, et al. The Canadian C-spine rule of radiology in alert and
stable trauma patients. JAMA. 2001;286:1841–8.
2. Hoffman JR, et al. Selective cervical spine radiography in blunt trauma: methodology of
the National Emergency X-Radiography Utilization Study (NEXUS). Ann Emerg Med.
1998;32(4):461–9.
3. Patel MB, Humble SS, Cullinane DC, Day MA, Jawa RS, Devin CJ, Delozier MS, Smith LM,
Smith MA, Capella JM, et al. Cervical spine collar clearance in the obtunded adult blunt trauma
patient: a systematic review and practice management guideline from the Eastern Association for
the Surgery of Trauma. J Trauma Acute Care Surg. 2015;78(2):430–41.
4. Selden NR, Quint DJ, Patel N, d’Arcy HS, Papadopoulos SM. Emergency magnetic reso-
nance imaging of cervical spinal cord injuries: clinical correlation and prognosis. Neurosurgery.
1999;44:785–92.
5. Chandra J, et al. MRI in acute and subacute post-traumatic spinal cord injury: pictorial review.
Spinal Cord. 2012;50(1):2–7.
276
ICU MANAGEMENT OF SPINAL CORD INJURIES
Catherine S. W. Albin and Sahar F. Zafar
TERMS
Spinal shock: The loss of muscle tone and areflexia in the acute period after spinal
injury before the onset of spasticity.
Neurogenic shock: The loss of vasomotor tone and sympathetic innervation to the
heart. Usually the result of lesions that are at or higher than T6, these injuries result
in peripheral vasodilation and an inability to produce reflexive tachycardia.
ADMISSION CHECKLIST
□□Ensure spinal precautions ordered, C-collar in place (for acute evaluation and
management of C-spine injuries, see page 273).
□□Evaluate the need for intubation if not already done for high lesions (>C5).
□□Emergent consult to orthopedics or neurosurgery (institution dependent) for
surgical evaluation
–– The Acute Spinal Cord Injury study demonstrated that those that undergo
decompressive surgery within 24 hours of injury were twice as likely to have
a two-grade ASIA Impairment Scale improvement at 6 months [1].
□□ Preventive monitoring for early complications of spinal injury:
–– Hypotension and bradycardia: Seen most commonly in patients with T6 or
higher lesions given the involvement of the sympathetic outflow tract. Both
may require conservative fluid administration, vasopressors (Phenylephrine
should be used with caution in T6 and higher lesions, as it may worsen bra-
dycardia) and atropine for bradycardia. Prudent to have external pacing pads
available for high T- and C-spine injuries.
○ MAP goal >85–90 for spinal perfusion should be considered and is
guideline-recommended (however note no randomized trials support this
practice). Duration varies although 5–7 days is used [2].
Respiratory insufficiency: Secondary to chest wall and diaphragm weak-
ness. Close monitoring of respiratory parameters like Negative Inspiratory
Force (NIF) and Vital Capacity (VC), as well as RR and pCO2.
○ Seen most frequently with C-spine injuries.
○ Incentive spirometry and chest physiotherapy is recommended for all
patients to prevent atelectasis and pneumonia.
–– Urinary retention: A Foley should be used for decompression in first days–
week after injury. Switch to intermittent catherization as soon as feasible to
reduce CAUTIs.
278
CLASSIC SYNDROMES
INJURY
LOCATION CLINICAL SYNDROME PICTURE
Central cord Greater loss of motor function in
syndrome the upper extremities compared Loss of motor
function Area of cord
spinal fracture/dislocation.
Cervical stenosis is a risk factor
279
PROGNOSTICATION IN SPINAL CORD INJURIES
The American Spinal Injury Association (ASIA) international standards is the pre-
ferred tool to standardize the exam and classify severity. It is available online.
–– Because of spinal cord swelling or spinal shock, the prognostic evaluation should
be deferred until the end of the acute hospitalization.
A systematic review [6] demonstrated that these factors effected neurologic recovery:
–– The severity of injury measured by the ASIA scale
–– Level of injury
–– The presence of a zone of partial preservation
The following factors affected functional outcome:
–– Severity of neurologic injury
–– Level of injury
–– Reflex pattern
–– Age
REFERENCES
1. Wilson JR, et al. Early versus late surgery for traumatic spinal cord injury: the results of a pro-
spective Canadian cohort study. Spinal Cord. 2012;50(11):840–3.
2. Ryken TC, Hurlbert RJ, Hadley MN, et al. The acute cardiopulmonary management of patients
with cervical spinal cordinjuries. Neurosurgery. 2013;72(Suppl 2):84–92.
3. Walters BC, et al. Guidelines for the management of acute cervical spine and spinal cord inju-
ries: 2013 update. Neurosurgery 2013; 60(CN_suppl_1):82–91.
4. Bracken MB. Steroids for acute spinal cord injury. Cochrane Database Syst Rev 2002;2.
280
5. Bracken MB, et al. Administration of methylprednisolone for 24 or 48 hours or tirilazad mesylate
for 48 hours in the treatment of acute spinal cord injury: results of the Third National Acute Spinal
Cord Injury Randomized Controlled Trial. JAMA. 1997;277(20):1597–604.
6. Wilson JR, Cadotte DW, Fehlings MG. Clinical predictors of neurological outcome, functional
status, and survival after traumatic spinal cord injury: a systematic review. J Neurosurg Spine.
2012;17(Suppl1):11–26.
7. Eldahan KC, Rabchevsky AG. Autonomic dysreflexia after spinal cord injury: systemic pathophys-
iology and methods of management. Auton Neurosci. 2018;209:59–70. https://doi.org/10.1016/j.
autneu.2017.05.002.
8. Karlsson AK. Autonomic dysreflexia. Spinal Cord. 1999;37(6):383–91.
281
MANAGEMENT OF THE POSTOPERATIVE CRANIOTOMY
PATIENT
Alison Paolino and Catherine S. W. Albin
CRANIOTOMY
• The bone is removed and put back on during the same surgical procedure.
CRANIECTOMY
• The bone is removed and kept off to relieve pressure and is put back on during a
later surgery called a cranioplasty.
A craniotomy is performed for various central nervous system (CNS) pathologies,
such as to clip an aneurysm, remove a tumor, or evacuate a hematoma. A craniec-
tomy is usually performed only to treat high intracranial pressures or prevent
herniation.
The postoperative treatment and expected postoperative ICU course will vary
significantly based on the reason for surgery. It is important for the ICU team to
understand the anatomy, surgical approaches, and what happened in the operating
room to anticipate, avoid, and manage postoperative complications.
CHECKLIST FOR ADMISSION
□□Collect vital information from surgical and anesthesia team (see above)
□□Document comprehensive neuro exam and confirm with surgical team any
changes from baseline
□□ Review postoperative imaging, if obtained
□□ Review and restart antiepileptic therapy, if indicated
□□ Define course for steroids or other ICP-control measures, note if patient was
on steroids before procedure (raising risk for adrenal insufficiency/hypotension)
284
The best way to prevent complications is by anticipating their probable occurrence
and developing strategies to deal with the complications when they occur.
285
GENERAL CRANIOTOMY POSTOPERATIVE COMPLICATIONS [1]
Tension Air in the cranium that is under pressure. May be located in any CNS
pneumocephalus compartment. Symptoms include headache, nausea, vomiting, seizures,
dizziness, obtundation. May see the “Mt Fugi Sign” on non-contrasted HCT
(two frontal lobe “peaks” surround by air). Requires urgent neurosurgical
evacuation via a new burr hole or insertion of drain via established burr hole.
CSF leak Most commonly seen with basilar skull fractures, transsphenoidal surgeries
(TSA), or posterior fossa craniotomy with dural opening. Clear fluid drains
through the skin incision, the eustachian tube (basilar skull fractures), or nose
and/or back of throat (TSA), especially if the patient leans forward. Puts the
patient at increased risk for infectious complications.
Diagnosis:
Infections Superficial infection often present earlier (1–2 weeks) and often can be treated
with debridement + systemic antibiotics. Deep infections often develop later
(>2 weeks) and may involve the bone flap (osteomyelitis). These infections
require surgical wound revision and removal of infected flap, as well as broad
spectrum coverage (gram positive, gram negative, and anaerobic).
Pseudomeningocele An abnormal collection of cerebrospinal fluid that occurs due to leakage from
CSF-filled spaces. Minor pseudomeningoceles can usually be followed. Large
collections may require a lumbar drain for CSF diversion.
OR positioning Depends on the surgical positioning, which should always be clarified with
complications anesthesia. Complications include compartment syndrome or peripheral nerve
entrapment syndrome. Laceration and pain may also be related to rigid
fixation of the skull during the operation.
286
Fig. 54.1 Significant tension pneumocephalus in a post-craniectomy patient. Suspected to have
resulted from air entrapment by malfunctioning shunt
Fig. 54.2 Diffuse pneumocephalus seen on CT scan in a patient that developed a CSF leak 1 week
after TSA
287
Special Case
Cerebral Angiogram & Craniotomy
During some craniotomies for vascular abnormalities, a cerebral angiogram is
performed intra-operatively in order to ensure the treatment aim has been achieved.
See page 291 for postoperative management specific to endovascular procedures.
Special Case
Sub-occipital/Retrosigmoid/Translabyrinthine Craniotomies
Used to access the cerebellopontine angle and cerebellum
□□May require EVD for obstructive hydrocephalus caused by post-operative
swelling infratentorially or to reduce pressure on healing dural incision
(thus lowering the risk of a CSF leak)
□□ Lower cranial nerve injuries may result in a high risk of aspiration and
dysphagia
□□ Fifth/seventh nerve injury may result in weakening of the corneal reflex,
leading to corneal ulceration (need eyedrops or eyelid taped shut during
recovery)
□□ Higher risk for CSF leaks, monitor incision site closely
□□ Usually complicated by nausea/vomiting neck muscle spasms, treat
aggressively
288
Special Case
Management Considerations for TSA/Pituitary Surgery Patients
Endocrine Considerations:
□□Post-operatively, high risk for Diabetes Insipidus (for management, see page
307) seen in 8–31% of patients, usually begins 24–48 hours after surgery
[2]; many patients will recover endogenous vasopressin secretion within
several days. Some will require lifelong exogenous ADH replacement. A
very small number of patients will have a “triple phase response” resulting
in a pattern of (1) transient DI, (2) SIADH, and then (3) permanent DI.
□□ Post-operatively, AM cortisol should be drawn for at least 2–3 mornings
after stress dose steroids have been stopped.
□□ AM Cortisol >450 nM = no concern for ACTH depletion. <100 nM =
requires baseline physiologic steroids; 100–450 = may have ACTH
depletion and likely require baseline steroids or at least stress dose
steroids during times of illness, endocrine should be consulted.
Special Monitoring:
□□Visual field testing
□□Asking specifically every morning about nasal drainage, a salty taste, fluid
cooling in the back of the throat – signs of a CSF leak
□□See above for management strategies for CSF leak.
Special Orders Needed for TSA Patients:
□□No positive pressure non-invasive ventilation
□□Sinus precautions
□□Consider pneumovax vaccine; if a CSF leak develops the patient is at high
risk for pneumococcal meningitis
□□Vasospasm and delayed cerebral ischemia has been described [3].
Postoperative transcranial doppler monitoring or CTAs can be considered.
REFERENCES
1. Kumar M, et al., editors. Neurocritical care management of the neurosurgical patient E-Book.
Elsevier Health Sciences; 2017.
2. Ricarte IF, et al. Symptomatic cerebral vasospasm and delayed cerebral ischemia following trans-
sphenoidal resection of a craniopharyngioma. J Stroke Cerebrovasc Dis. 2015;24(9):e271–3.
3. Hensen J, Henig A, Fablbush R, Meyer M, Boehnert M, Buchfelder M. Prevalence, predictors
and patterns of postoperative polyuria and hyponatremia in the immediate course after transs-
phenoidal surgery for pituitary adenomas. Clin Endocrinol. 1999;50(4):431–9.
289
POSTOPERATIVE MANAGEMENT
OF CEREBROVASCULAR PATIENTS
Alison Paolino and Catherine S. W. Albin
The same information should be collected as for other post-operative patients (Chap.
54), with the following additions:
□□ Any injury to catheterized arteries
□□ Excess bleeding from access site Postoperative Angiogram Checks
□□ Particularly important to clarify □□
Vital signs and neuro assess-
blood pressure goals ment at least Q2H
□□
Neurovascular and pulse checks
Postoperative Complications to
Monitor for:
□□
Groin (or access site) checks
–– Q15 minutes × 4
□□ Stroke –– Q30 minutes × 4
□□ Injury to catheterized arteries –– Q1 hour × 3
□□ Allergic reaction to the contrast
dye and other medicines used
in the procedure
□□ Hematoma and/or pseudoaneurysm development at access site, placing
patient at risk for arterial thromboembolic complications (monitoring for cold/
pulseless feet (groin access) or hand (radial access site)). Retroperitoneal
hematomas, arteriovenous fistula, arterial occlusion, femoral neuropathy, and
infection are all far less common access site complications.
292
COMMONLY TREATED NEUROVASCULAR PATHOLOGIES (Continued)
CEREBRAL
ARTERIOVENOUS
CEREBRAL MALFORMATION CAVERNOUS MOYA-MOYA
ANEURYSM (AVM) MALFORMATION DISEASE
Procedure Craniotomy for Craniotomy for Craniotomy for Craniotomy for
clip ligation vs. resection of resection. superior temporal
endovascular AVM. Many can These are artery (STA) to
coiling, web, or also be treated with angiographically MCA (direct
flow diverter to endovascular silent lesions. bypass)
prevent rupture embolization in
or re-rupture conjunction with Craniotomy for
radiosurgery or Encephaloduroar-
See page 241 resection teriomyosynangio-
for details sis (EDAMS)
regarding (indirect bypass)
treatment
strategies and
229–244 for
SAH
management
Intraoperative Intraoperative Intraoperative Depending on the Intraoperative
monitoring cerebral cerebral angiogram, location cerebral
angiogram SSEPs, and/or neuromonitoring angiogram
intraoperative EEG may be used.
may be used
Major Rupture or Hemorrhage or Ischemia/ Rupture or
intraoperative re-rupture of re-hemorrhage of Hemorrhage re-rupture of
complications aneurysm; AVM; ischemia fragile arteries
ischemia from Seizures leading to
clip migration Note that DMSO, if cerebral
or coil prolapse used, can induce Damage to hemorrhage
(endovascular) vasospasm, surrounding
[1] angionecrosis, structures Ischemia
arterial thrombosis,
Peri-procedural and vascular rupture Peri-procedural
vasospasm [2] vasospasm
293
COMMONLY TREATED NEUROVASCULAR PATHOLOGIES (Continued)
CEREBRAL
ARTERIOVENOUS
CEREBRAL MALFORMATION CAVERNOUS MOYA-MOYA
ANEURYSM (AVM) MALFORMATION DISEASE
Notable Ischemia Normal Venous infarct if a MMD patients
postoperative perfusion developmental have chronically
complications Seizures pressure venous anomaly deranged CBF
breakthrough (DVA) is disrupted and CVR which
Complications (NPPB): Restoration during surgery puts them at risk
of rerupture, if of normal perfusion for:
occures after AVM resection Brainstem cavernous
results in increase in malformations often Ischemia/
Peri-procedure arterial flow to lay adjacent to hypoperfusion
vasospasm adjacent areas and critical structures, (can result from
tissue that has and resection may the failure of CVR,
theoretically been result in cranial hypoperfusion
deprived of normal nerve palsies, due to competition
vascular ataxia, spasticity, between graft and
autoregulation. May and swallowing native collaterals,
led to hyperemia, difficulties graft occlusion).
edema, and COSS trial 15%
potentially of patients had
ICH. Treated by perioperative
conservatively stroke [3]
lowering patients’
SBP for 24 hours Cerebral
post-procedure hyperperfusion
syndrome
ICH may also result (CHS): similar to
from can use NPPB, occurs due
micro-perforation of to rapid increase
the vessels or in blood flow to
thrombosis of the chronically poorly
adjacent venous autoregulated/
system resulting in ischemic regions
occlusive hyperemia of the brain
294
COMMONLY TREATED NEUROVASCULAR PATHOLOGIES (Continued)
CEREBRAL
ARTERIOVENOUS
CEREBRAL MALFORMATION CAVERNOUS MOYA-MOYA
ANEURYSM (AVM) MALFORMATION DISEASE
Vascular SBP goal − SBP ~10–20% Goal normotension Antiplatelets are
craniotomy- determined lower than baseline to prevent commonly used to
specific based on if the to prevent NPPB hemorrhage maintain graft
postoperative aneurysm is − Normovolemia patency
orders considered fully
secured Some centers may
seeak a
Note that MAP>90–100 (or
flow-diverted slightly above
aneurysms are baseline) for at
not considered least 24 hours for
secured until cerebral
endothelization perfusion.
which is not
completed in Avoid
the acute phase compressing
of management donor side of
graft (such as with
a CPAP or
tight-fitting nasal
canula)
Misc. See page 235 ~7% AVMs have Most supratentorial; The late
for details on associated flow- 9–35% infratentorial development of a
management of related aneurysms SDH or an EDH
aneurysm- [4] are two late
associated SAH complications to
be aware of [5]
295
Fig. 55.1 Spetzler-Martin grade 4 right parietal paramedian arteriovenous malformation primarily
supplied by the distal anterior cerebral artery and branches of PCA with superficial venous drainage
into the superior sagittal sinus and superficial venous drainage in the cortical veins and vein of
Labbe. Deep venous drainage is noted into the distal vein of Galen
296
Fig. 55.2 T2 FLAIR MRI sequence showing a cavernous malformation in the pons with internal hem-
orrhage products of different ages
REFERENCES
1. Brisman JL, Song JK, Newell DW. Cerebral aneurysms. N Engl J Med. 2006;355(9):928–39.
2. Chaloupka JC, et al. A reexamination of the angiotoxicity of superselective injection of DMSO in
the swine rete embolization model. Am J Neuroradiol. 1999;20(3):401–10.
3. Powers WJ, et al. Extracranial-intracranial bypass surgery for stroke prevention in hemo-
dynamic cerebral ischemia: the Carotid Occlusion Surgery Study randomized trial.
JAMA. 2011;306(18):1983–92.
4. Stapf C, et al. Concurrent arterial aneurysms in brain arteriovenous malformations with haemor-
rhagic presentation. J Neurol Neurosurg Psychiatry. 2002;73(3):294–8.
5. Andoh T, et al. Chronic subdural hematoma following bypass surgery—report of three cases—.
Neurol Med Chir. 1992;32(9):684–9.
297
PREPARATION FOR BRAIN DEATH TESTING
Catherine S. W. Albin and Sahar F. Zafar
Hospital policies differ on the exact testing procedures, timing, the requirement of
who can declare, and need for multiple examiners [1]. Always print out and exactly
follow the hospital’s policy. To help, the AAN has an easily accessible checklist/
worksheet based on their guidelines [2] that is free and available online by searching
“AAN brain death guidelines.”
One important element that frequently delays testing or erroneously prevents
testing is the perception that the patient is breathing over the ventilator.
Patients that appear to meet criteria for brain death but are “triggering” the
vent should be assessed for auto-triggering the ventilator.
VENTILATOR AUTOTRIGGERING & BRAIN DEATH
A brain-dead patient can appear to have respiratory effort due to the cardiopulmo-
nary consequences of brain death and the hyperdynamic state that often accompa-
nies this syndrome leading to [3–5]:
Mvmt of air w/
Displacement in "Breath" in the
A hyperdynamic each cardic cycle
intrathoracic contents absence of
precordium triggers the vent
w/ each cardiac cycle respiratory drive
(more sensitive)
Other important steps to ensure are completed prior to brain death and apnea
testing:
□□ Confirm that the patient has an explained and neuroradiographic evident cause
of irreversible coma (not just in a locked-in state or deep coma as may be the
case with a devastating brainstem injury).
□□ The AAN Guidelines do not specify if there must be evidence of diabetes
insipidus (pituitary death), but legal policy requires more than just “brainstem”
death and states “whole brain death.” If there is any uncertainty if the patient
qualifies, seek expert guidance.
□□ Patients with facial injuries and/or baseline cranial nerve deficits will need
confirmatory testing and cannot be declared with clinical examination alone.
□□ Review the medication list: all sedatives and paralytics need to be off for at
least five half-lives and may need longer in patients who have been treated with
therapeutic hypothermia. Train of four should be used to confirm the patient is
not paralyzed. Work with unit pharmacist to determine the appropriate time for
testing. In patients with suspected drug overdoses or intoxications, serial urine
toxicology or serum toxicology screens may be appropriate or ancillary testing
should be used in conjuction with clinical testing. Seek expert guidance.
□□ Screen labs for major metabolic derangements such as uremia and hyperam-
monemia; mild hypernatremia is permissible.
□□ Screen for endocrine abnormalities such as hypothyroidism that might con-
found the exam.
□□ Optimize the patient hemodynamically. SBP should be ≧ 100 mmHg.
–– Diabetes insipidus is common in herniation and may result in profound hemo-
dynamic collapse. Ensure this is treated. See page 307.
–– Pressors are okay.
□□ Patient must be ≧ 36°C.
300
For Apnea Testing:
□□ Patient must be normocapnic (paCO2 35–45 mmHg unless baseline CO2 is
suspected to be higher; hospitals have different policies on how this should be
handled).
□□ Patient should be pre-oxygenated for a goal PaO2 > 200 mmHg.
□□ Arterial line for hemodynamic monitoring and blood gas sampling.
□□ Pressors should be available – ideally in the room and already in line – as
progressive acidemia will lead to vasodilation and often hypotension.
Once these requirements are met, the patient is ready for bedside testing.
Supplies you will need:
□□A bright pen light
□□Tongue depressor to check gag
□□Q-tip to check corneal reflex
□□50 cc of ice-cold water in a syringe with a tubing attachment that can go into
the ear canal (x2)
□□Tracheal suctioning supplies if there is no in-line suctioning
□□An insufflation catheter for apnea testing (respiratory therapists should be
available to help with the apnea test)
When doing brain death testing, it is incredibly important to document
precisely.
ANCILLARY TESTING
Ancillary testing must be completed if clinical examination cannot be fully performed
or if the apnea testing is aborted. This can be done with a cerebral angiogram,
nuclear medicine 99mTc-HMPAO SPECT, or TCD test. EEG is no longer preferred.
MRA, CTA, and SSEPs are not accepted. If an ancillary test is needed, speak to the
technologists and interpreting physicians ASAP, as these tests may require special
credentialing for interpretation and often require some coordination.
301
REFERENCES
1. Greer DM, et al. Variability of brain death determination guidelines in leading US neurologic
institutions. Neurology. 2008;70(4):284–9.
2. Wijdicks EFM, et al. Evidence-based guideline update: determining brain death in adults: report
of the Quality Standards Subcommittee of the American Academy of Neurology. Neurology.
2010;74(23):1911–8.
3. Arbour R. Cardiogenic oscillation and ventilator autotriggering in brain-dead patients: a case
series. Am J Crit Care. 2009;18(5):496–88.
4. Wijdicks EFM, Manno EM, Holets SR. Ventilator self-cycling may falsely suggest patient effort
during brain death determination. Neurology. 2005;65(5):774.
5. Cole RP. Cardiogenic oscillations and apparent ventilation in suspected brain death.
Resuscitation. 2003;56(3):335.
302
NUTRITION IN THE NEUROICU
Carmen Lo
GOALS
• Feed early: In patients who are not in shock, start patients on tube feeds (TF)
within 24–48 h (even only 10 ml/h if unable to advance rate). This helps establish
feeding tolerance and benefits GI integrity and immune response.
• Meet 80–100% nutritional needs by ICU days 3–7 for optimal clinical outcome:
° Achieving 80% energy need and close to 100% protein need (1.2 g/kg protein) in
48–72 h correlates with improved mortality in ICU patients [1] especially for BMI
>30, BMI <18.5, and NUTRIC score* >5 populations.
° TBI patients: early enteral nutrition promotes neurologic recovery [2]. One study
reported that every 10 kcal/kg increase of energy intake during the first 5 days
(up to 25 kcal/kg) reduced the 2-week post-injury mortality by 30–40% [3].
*NUTRIC score: score to quantify the risk of critically ill patients developing
adverse event that may be modifiable by aggressive nutrition therapy; variables
include age, Apache II, SOFA, number of comorbidities, days from hospital to
ICU admission, and IL-6.
• Avoid underfeeding/overfeeding:
° Underfeeding can worsen the patient’s nutritional status and compromise clinical
outcome. Limit interruption and holding of feeding as able.
–– Compensatory feeding helps make up the difference when TF interruption is
needed. If a protocol like this exists in the institution, then the RN can adjust
TF rate up to a maximum of ~150 ml/h to catch up the daily TF goal volume.
–– When pursuing compensatory feeding, check the gastric residual order
(>200 ml: start promotility agent; >500 ml: stop TF) which helps deliver TF
safely and reduces unnecessary holding.
° Overfeeding can lead to increased oxygen requirement/ventilator dependency,
blood sugar/insulin requirement, electrolyte derangement, and GI burden, which
can contribute to the worsening of clinical outcome. The ICU RD (registered
dietitian) can help you assess patient and customize TF goal to best match
patient needs in different phases of ICU care.
° For patients on IV medication in lipid emulsions such as propofol (1.1 kcal/ ml)
and clevidipine (2 kcal/ml), seek RD guidance to avoid under-/overfeeding.
• Factors that might influence tube feeding strategy: BMI, pre-admission nutritional
status, renal function, blood glucose, and volume status.
TIPS
Food-drug interactions: Certain medications (such as levothyroxine and phenyt-
oin) require holding of TF 1 hour before and after medication administration. Work
with the RD to adjust TF order with a compensated rate for the shortened daily TF
infusion time.
Electrolyte derangements (especially hypophosphatemia): Common in malnour-
ished patients who are at high risk of refeeding syndrome (though this can also occur
in patients with normal nutritional status). Nutritional support can exacerbate electro-
lyte derangements and therefore requires close electrolyte monitoring and aggres-
sive repletion. Phos repletion goal ≥2.6.
• If Phos <1.5: Consider holding TF. Replete and recheck ~2 hours after
replacement.
• If Phos 1.5–2.5: Can start/continue TF.
• Recommend BID Phos check until TF is at goal and Phos is stable.
• Replete with IV Phos (15–45 mmol × 1) if Phos <1.8 or repeatedly low.
• Replete with PO/Enteral Phos NAK (1 pkt QID ~32 mmol) if Phos ≥1.8.
Blood sugar management when on tube feeds: Insulin requirements for patients
with hyperglycemia may change as TF are adjusted. Upon new TF recommendation
for changes, RD will notify the responding clinician on the difference of carbohydrate
provision.
304
APPROX ML/H GOAL FOR
FIRST 24–48 H (BASED
CALORIE ON AVERAGE-SIZED
FORMULA COUNT ADVANTAGES PERSON, ABOUT ~70 KG)
Osmolite 1.06 cal/mL Isotonic. Easy to tolerate, easily absorbed. 50 mL/h (total calorie
1 cal No fiber. A good default choice unless there 1272)
are factors that make an alternative choice
better
Osmolite 1.5 cal/mL Concentrated. Advantageous in situations 35 mL/h (total calorie
1.5 where lower water balance is preferred: 1260)
Need for negative TBB and SIADH. Can
also be used if there is a high energy need.
Not suggested for patient with high
refeeding risk due to high caloric density
Promote 1.0 cal/mL Higher protein formulation. Best for obese 50 mL/h (total calorie
patients (BMI > 30) or those on high-dose 1200)
propofol for an extended period
Nepro 1.8 cal/mL Best for end-stage renal patients where 30 mL/h (total calorie
volume overload or electrolytes have been 1296)
difficult to manage (i.e., Phos and K are
high, and there is low UOP). Many patients
with mild CKD/AKI can be managed with
Osmolite
Glucerna 1.0 cal/mL Low in carbs but high in fat (which may slow 50 mL/h (total calorie
down gastric emptying). Not the preferred 1200)
choice of ICU patient, not optimal for
long-term feeding but can be used
adjunctively with insulin for patients with
refractory hyperglycemia
Jevity 1.0 cal/mL Fiber-containing formula. Can be used for 50 ml/h (for Jevity
and bowel management in well-established 1.0 cal/mL)
1.5 cal/ mL tube-feeding patient. Helps liquid stool at or
times. Avoid when patient is 35 ml/h (for Jevity
hemodynamically unstable or with distended 1.5 cal/mL)
abdomen
305
REFERENCES
1. Heyland DK, et al. Identifying critically ill patients who benefit the most from nutrition therapy: the
development and initial validation of a novel risk assessment tool. Crit Care. 2011;15(16):R268.
2. Taylor SJ, et al. Prospective, randomized, controlled trial to determine the effect of early
enhanced enteral nutrition on clinical outcome in mechanically ventilated patients suffering head
injury. Crit Care Med. 1999;27(11):2525–31.
3. Hartl R, et al. Effect of early nutrition on deaths due to severe traumatic brain injury. J Neurosurg.
2008;109(1):50–6.
306
HYPERNATREMIA IN THE NEUROICU
Melissa Bentley and Catherine S. W. Albin
HYPERNATREMIA
IATROGENIC FREE WATER DEFICIT DIABETES INSIPIDUS (DI)
Etiology Most commonly, From lack of fluid Commonly seen:
from hypertonic administration during − In herniation
saline hospitalization, − After transsphenoidal surgery (if
administration impaired thirst disruption of the pituitary stalk)
mechanism, − With pituitary tumors or
insensible losses, GI inflammation of the pituitary
losses, and diuretics gland (such as those seen with
sarcoidosis)
− Pituitary stalk compression
− Pituitary/hypothalamus damage
Pathophysiology Salt administration Free water loss via Decreased anti-diuretic hormone
the kidney and gut or (ADH) production from damage to
insensibly with the pituitary gland results in
impaired mechanism excessively large dilute urine output
to replenish free
The rapid loss of dilute urine leads
water
to a very abrupt and significant
increase in serum sodium levels.
Monitor Monitor serum Monitor serum *() denotes the findings that are
sodium sodium; stricts I/O concerning for uncontrolled DI*
recording − Urine output (usually >200 cc ×
2 hours in the absence of a large
fluid intake)
− Serum Na (often >145 mg/dL)
− Serum Osm (>290 mmol/L)
− Urine Osm (<200 mOsm/kg)
Signs/ Thirst Thirst Polydipsia, polyuria, tachycardia,
symptoms hypotension
Complications Acute kidney Acute kidney injury Hypotension, hemodynamic
injury (may be (secondary to low collapse, and death (if uncorrected)
related to the volume status and
metabolic acidosis prerenal AKI)
caused by the
concurrent
administration of
chloride)
–– The free water deficit (which is based on serum sodium and weight) can be cal-
culated using online calculators like Nephromatic.com.
–– Note that in the NeuroICU many patients have cerebral edema, and rapid shifts
in sodium could result in worsening cerebral edema and herniation. As such,
D5W is not routinely used to correct hypernatremia in the NeuroICU.
–– Whenever possible, free water should be administered enterally.
–– If the patient has hypovolemia hypernatremia, correcting volume status will often
slowly correct the sodium.
–– If the patient is hypervolemic, consider diuresis with an agent that promotes
natriuresis such as metolazone, hydrochlorothiazide, or chlorothiazide (which
inhibit sodium reabsorption at the distal tubule).
Note II: These are very closely related to desmopressin = DDAVP; these compounds
are slightly modified to have less vasoactive properties (ie. they are not pressors).
Management:
□□Admit to ICU for dose finding.
□□Hourly fluid I&Os.
□□At least Q4H Uosm.
□□Frequent serum sodiums (Q4H–Q6H is appropriate initially).
□□Given frequent blood draws, consider placing an arterial line.
Treatment:
**Serum sodium is a reflection of what “has” happened; Urine Osm is a reflection of
how management is “currently” going.**
–– In a situation where the patient is at risk for DI, consider administration of anti-
diuretic hormone when urine output is >200 cc/hour × 2 hours and Urine
Osm 100–200 mmol/L – no need to wait for sodium to rise.
–– When this threshold is met, vasopressin 2.5–5 units IV × 1 (although institutional
thresholds and doses may vary) should be administered, while an infusion is
being prepared. This bolus dose of vasopressin has an effect for 4–6 hours.
–– Begin vasopressin infusion at 0.5–1 units/hour and titrate to a urine osmolarity of
300–500 mmol/L.
–– The goal is that intake should match output. If the patient is awake, they should
be encouraged to drink to thirst. If they are not, replace lost fluid with an isotonic
308
solution like Plasm-A-lyte (hypotonic solutions like LR are generally avoided in
brain injuried patients).
–– If sodium is rising despite the infusion and administration of crystalloid, consider
either an additional bolus of vasopressin or increasing the drip rate or increasing
the amount of free water given in the gut. D5W is typically avoided in patients
with severe brain injuries, but there are some situations were this may be
appropriate.
Converting to oral DDAVP:
–– There is variability in the absorption of oral DDAVP; thus conversion from vaso-
pressin to DDAVP is not an exact science.
–– Generally, try either 0.1 or 0.2 mcg of oral DAVP and watch Uosm and urine
output. If urine output remains high, a higher dose is required. When urine osmo-
larity begins to fall 100 mmol/L × 2 checks, the patient probably requires
another dose.
–– Generally the effect lasts between 8 and 24 hours. Some patients may require
very high initial doses and then slowly be weaned to just a nighttime doses.
–– If exceedingly high doses are required, consider switching to subcutaneous or
intranasal spray formulations which have much more consistent absorption and
are thus order of magnitudes more effective. Intranasal sprays should not be
used in post-TSA patients.
–– Note that concurrent glucocorticoid administration may require an increased
DDAVP dose.
309
HYPONATREMIA IN THE NEUROICU
Catherine S. W. Albin and Sahar F. Zafar
Hyponatremia
Lab Findings:
Lab Findings: UOsm>100 mosmol/kg
Lab Findings: UNa<10 mEq/L UOsm>100 mosmol/kg UNa (often)>25 mmol/L
FeNa< 1% UNa>25 mmol/L
(Note that renal losses, extra-renal losses, and primary polydipsia are also etiologies of hypotonic
hyponatremia, but as these are less common in the neuroICU population, they are not covered here.)
Treatment
–– In patients not at risk for vasospasm or who can tolerate a net negative fluid
balance: fluid restriction and increase solute intake; in extreme cases
a vaptan drug can be considered.
–– In patients at risk for vasospasm or who otherwise cannot be volume restricted:
° Hypertonic saline (3% infusion often used)
° Salt tabs (sample dose ~1–2 g TID may cause nausea).
° Oral urea 15 g QD to Q8H [1].
° Fludrocortisone should not be used as it promotes sodium retention and water
retention. But, because ADH is already over-secreted, this can result in water
being retained more than sodium and worsen hyponatremia.
Treatment
–– Salt replacement. Maintain even to slightly positive fluid balance.
–– Volume replacement with normal saline.
–– Can consider salt tabs to reduce number of fluid boluses.
–– Can trial fludrocortisone which promotes sodium retention. When used, closely
monitor for hypokalemia [2].
REFERENCES
1. Soupart A, et al. Efficacy and tolerance of urea compared with vaptans for long-term treatment of
patients with SIADH. Clin J Am Soc Nephrol. 2012;7(5):742–7.
2. Misra UK, Kalita J, Kumar M. Safety and efficacy of fludrocortisone in the treatment of cerebral
salt wasting in patients with tuberculous meningitis: a randomized clinical trial. JAMA Neurol.
2018;75(11):1383–91.
312
PRESSORS AND INOTROPES COMMONLY USED
IN THE NEUROICU
Catherine S. W. Albin and Megan E. Barra
DOSE
DRUG MECHANISM BEST FOR (MCG/KG/MIN)A MONITOR FOR
Norepinephrine α > > β1 > β2: Vasodilatory 0.01–3 mcg/kg/min Tachycardia,
(Levophed) Results in shock, (~0.5–150 mcg/min) bradycardia,
− Vasoconstriction cardiogenic arrhythmias,
− Increases cardiac shock, ischemia, severe
contractility neurogenic hypertension (pts
shock on B-blockers)
Phenylephrine Alpha only: Vagally 0.1–9 mcg/kg/min Bradycardia,
(Neosynephrine) − Potent mediated or (~20–500 mcg/min) severe peripheral
vasoconstriction medication- and visceral
− Minimal effect on induced vasoconstriction
HR, may cause hypotension
Avoid in
reflex bradycardia
cardiogenic
shock, increases
afterload or
neurogenic shock
T5 and above
REFERENCES
1. Overgaard CB, Dzavík V. Inotropes and vasopressors: review of physiology and clinical use in
cardiovascular disease. Circulation. 2008;118(10):1047–56.
2. Lannes M, et al. Milrinone and homeostasis to treat cerebral vasospasm associated with
subarachnoid hemorrhage: the Montreal Neurological Hospital protocol. Neurocrit Care.
2012;16(3):354–62.
314
SEIZURE PROPHYLAXIS IN THE NEUROICU
Amanda Rivera, Stephanie Seto, and Megan E. Barra
ROUTINE
PROPHYLAXIS
INDICATION INCIDENCE INDICATED RECOMMENDATIONS
Traumatic brain 4–42% − Administer empiric prophylaxis for
injury (TBI) [1] 7 days post-injury
− More beneficial in early vs. late PTS
− Agents: Preferred – levetiracetam, phenytoin
Aneurysmal 1–18% − Administer empiric prophylaxis until
subarachnoid aneurysm is secured
hemorrhage (aSAH) − Short-course (3–7 days) prophylaxis
[2] preferable if indicated
− Phenytoin is not recommended routinely for
seizure prophylaxis
Brain neoplasm [3] 10–45% −R
outine prophylaxis not recommended as
shown to be ineffective in preventing first
seizure and have potential side effects
− No benefit shown in patients undergoing
supratentorial meningioma resection or in
metastatic brain tumors
− A short course may be indicated
postoperatively in patients presenting with
seizures
Intracerebral 5.5–24% − Insufficient evidence to support the use of
hemorrhage [4, 5] prophylactic AEDs
Ischemic stroke [3] 4–23% −R
isk factors: hemorrhagic conversion,
cortical involvement, involvement of >1 lobe
− Insufficient evidence to support the use of
prophylactic AEDs
Postoperative 15–20% − L imited evidence to support the prophylactic
craniotomy [3] use of AEDs in post neurosurgery patients
− Levetiracetam preferred over phenytoin
(due to lower ADEs)
Vascular lesions [3] Variable − Insufficient evidence to support the use of
prophylactic AEDs
Cerebral venous Up to 40% − Insufficient evidence to support the use of
thrombosis (CVT) [3] prophylactic AEDs
REFERENCES
1. Yerram S, Katyal N, Premkumar K, Nattanmai P, Newey CR. Seizure prophylaxis in the neurosci-
ence intensive care unit. J Intensive Care. 2018;6(1):17.
2. Carney N, Totten AM, O’Reilly C, Ullman JS, Hawryluk GW, Bell MJ, Bratton SL, Chesnut R,
Harris OA, Kissoon N, Rubiano AM. Guidelines for the management of severe traumatic brain
injury. Neurosurgery. 2017;80(1):6–15.
3. Gilmore EJ, Maciel CB, Hirsch LJ, Sheth KN. Review of the utility of prophylactic anticonvulsant
use in critically ill patients with intracerebral hemorrhage. Stroke. 2016;47(10):2666–72.
4. Hemphill JC III, Greenberg SM, Anderson CS, Becker K, Bendok BR, Cushman M, Fung
GL, Goldstein JN, Macdonald RL, Mitchell PH, Scott PA. Guidelines for the management of
spontaneous intracerebral hemorrhage: a guideline for healthcare professionals from the
American Heart Association/American Stroke Association. Stroke. 2015;46(7):2032–60.
5. Diringer MN, Bleck TP, Hemphill JC, Menon D, Shutter L, Vespa P, Bruder N, Connolly ES,
Citerio G, Gress D, Hänggi D. Critical care management of patients following aneurysmal sub-
arachnoid hemorrhage: recommendations from the Neurocritical Care Society’s Multidisciplinary
Consensus Conference. Neurocrit Care. 2011;15(2):211.
316
VENOUS THROMBOEMBOLISM PROPHYLAXIS
IN THE NEUROICU
Stephanie Seto and Megan E. Barra
PHARMACOLOGIC AGENTS
USUAL
DRUG DOSING DOSE ADJUSTMENTS CONSIDERATIONS
Unfractionated 5000 units SQ Obesity (e.g. BMI > 40 kg/m2, The q8h strategy is preferred
heparin q12h or q8h weight > 150 kg): Consider in trauma patients
7500 units SQ q8h
Renal dysfunction: No dose
adjustment required
Enoxaparin 40 mg SQ Obesity (e.g. BMI > 40 kg/m2, More frequent dosing such as
q24h weight > 120 kg): Consider 30 mg SQ q12h may be
40 mg SQ q12h if normal better for trauma and spinal
renal function cord injury patients
Low body weight (<50 kg):
Consider 30 mg SQ q24h
Renal dysfunction:
− CrCl 15–29 mL/min consider
30 mg SQ q24h (or use UFH)
− CrCl <15 mL/min use UFH
− Avoid use if fluctuating renal
function or high bleed risk
318
INCIDENCE TIMING OF VTE PREFERRED
INDICATION OF VTE INITIATION RECOMMENDATIONS AGENT
Brain tumor Up to 31% As soon as feasible − Initiate pharmacologic LMWH
prophylaxis upon
hospitalization if low risk
of major bleeding and
lack signs of hemorrhagic
conversion
− May consider
combination
pharmacologic/
mechanical prophylaxis if
high risk
Spinal cord Up to 80% As soon as feasible, − Pharmacologic LMWH preferred.
injury within 72 h of prophylaxis as soon as When UFH used
injury bleeding is controlled q8h preferred
− If pharmacologic over q12h
prophylaxis is not
possible, initiate IPC
− Mechanical prophylaxis
alone not recommended
if can tolerate
pharmacologic agent
Neuromuscular 3–7% As soon as feasible − Pharmacologic Unknown/
disease during acute prophylaxis preferred insufficient
hospitalization over IPCs/GCS but can evidence
use mechanical methods
when risk of bleeding
deemed high
REFERENCES
1. Nyquist P, Bautista C, Jichici D, Burns J, Chhangani S, DeFilippis M, Goldenberg FD, Kim K, Liu-
DeRyke X, Mack W, Meyer K. Prophylaxis of venous thrombosis in neurocritical care patients: an
evidence-based guideline: a statement for healthcare professionals from the Neurocritical Care
Society. Neurocrit Care. 2016;24(1):47–60.
2. Sherman DG, Albers GW, Bladin C, Fieschi C, Gabbai AA, Kase CS, O’Riordan W, Pineo GF,
PREVAIL Investigators. The efficacy and safety of enoxaparin versus unfractionated heparin for
the prevention of venous thromboembolism after acute ischaemic stroke (PREVAIL Study): an
open-label randomised comparison. Lancet. 2007;369(9570):1347–55.
319
3. CLOTS (Clots in Legs Or sTockings after Stroke) Trials Collaboration. Effectiveness of
intermittent pneumatic compression in reduction of risk of deep vein thrombosis in patients
who have had a stroke (CLOTS 3): a multicentre randomised controlled trial. The Lancet.
2013;382(9891):516–24.
4. Sauro KM, Soo A, Kramer A, Couillard P, Kromm J, Zygun D, Niven DJ, Bagshaw SM, Stelfox
HT. Venous thromboembolism prophylaxis in neurocritical care patients: are current practices,
best practices? Neurocrit Care. 2019;30(2):355–63.
5. Viarasilpa T, Panyavachiraporn N, Jordan J, Marashi SM, Van Harn M, Akioyamen NO, Kowalski
RG, Mayer SA. Venous thromboembolism in Neurocritical care patients. J Intensive Care Med.
2019;7:0885066619841547.
320
PART V
IMPORTANT REFERENCES
BRAINSTEM ANATOMY
Catherine S. W. Albin and Sahar F. Zafar
Midbrain
Cerebral Peduncle;
with CST
Red Nucleus
Substantia Nigra
Medial Longitudinal
Fasciculus
Oculomotor
nucleus
ML+
Spinothalamic tract
Mesencephalic nucleus of the
PAG trigeminal nerve
Cerebral Aquaduct
Pons
Basilar
Artery
CST CN VI
ML
CN VII
Spinothalamic tract
CN VIII
Trigeminal Nucleus
Cochlaear
MLF Nucleus
Facial Vestibular
colliculus VII nucleus Nuclei
4th Ventricle PPRF & VI
Inferior Cerebellar
Nucleus
Peduncle
MLF = medial longitudinal fasciculus
CST = spinothalamic tract
ML = medial lemniscus
CST
Vertebral Artery
CN XII
Inferior Olivary Nuclei
ML
CN X
Spinothalamic Tract
Nucleus Ambiguus
324
NEUROICU INTRAVENOUS FLUID COMPOSITIONS
Megan E. Barra
326
ANTI-SEIZURE MEDICATION CHART FOR USE IN ADULTS
Megan E. Barra and David Fischer
328
OTHER (E.G.,
MONITORING,
SIDE EFFECTS DRUG
SEIZURE (COMMON, RARE METABOLISM/ INTERACTIONS,
NAME TYPE MECHANISM PO DOSING IV DOSING BUT BAD) BINDING LEVEL INFO PHARMACOKINETICS PREGNANCY)
Clobazam, CLB Broad Benzodiazepine, Initial: 5 mg BID. Aggression, Metabolism: hepatic Clobazam: T ½: 36–42 hr Dec’s OCPs
(OnfI) spectrum, inc freq of GABA Adjust: Inc irritability, fever, Enzyme: CYP3A4, 30–300 ng/mL T ½ of active (weak). Inc’d by
Lennox- qweekly up to excessive salivation, 2C19 Norclobazam: metabolite: 71–82h CBD
Gastaut 20 mg BID URI Active metabolite. 300–3000 ng/ Tpeak: STerat/CTerat
syndrome (10mg for Inhibitor: CYP2D6 mL 0.5–4 hr(tab) unknown, BF
weight < 30 kg) (weak) Not routinely 0.5–2 hr(susp) unknown (in milk).
Dose adjust: Inducer: CYP3A4 measured Max onset: 5–9 days Risk of neonatal
hepatic (weak) withdrawal
Protein binding: ~80%
Clonazepam, Adjuvant for Benzodiazepine, Initial: 0.5–1 mg Paradoxical Metabolism: hepatic 15–70 ng/mL, T ½: 17–60 hr Dec’d by
CZP (Klonopin) myoclonic inc freq of GABA TID. aggression and Enzyme: CYP3A4 though not well Tpeak: 1–4 hr PHT. Inc’d by VGB
and atonic receptor Can dec to QD, anxiety established Onset: 2–40 min STerat
seizure Cl- channel or inc qweekly low-moderate,
opening up to 20 mg/ CTerat unknown,
day. Can inc BF unknown (in
quickly as milk). Risk of
inpatient neonatal
withdrawal
Diazepam, DZ Abortive for Benzodiazepine, 2–10 mg For sz <5 Hypotension, Metabolism: hepatic T ½: 33–45 hr, STerat/CTerat
(Valium) prolonged or inc freq of GABA BID-QID min: 0.15 respiratory Enzyme: CYP3A4, prolonged with multiple unknown, BF
cluster receptor mg/kg IV (5 depression 2C19 doses unknown (in milk).
seizures Cl- channel mg/min) or Paradoxical Active metabolites: T½ CNS:15–20 min Risk of neonatal
opening PR up to aggression and N-desmethyldiazepam T ½ of active withdrawal
10 mg per anxiety and temazepam which metabolite: 100 hr
dose (5 mg/ then metabolize to Tpeak: 1 min (IV)
min), repeat oxazepam 15 min–2hr (PO)
q5 min Onset: 1–3 min(IV),
2–10 min (PR)
Eslicarbazepine Focal Na+ channels Initial: 400 mg Nausea Metabolism: hepatic, T ½: 13–20 hr Na, LFTs before
ESL (Aptiom) seizures (can QD. Rash, SJS, PR UGT Prodrug for active Tpeak: 1–4 hr starting and on
worsen Inc by prolongation, monohydroxy maintenance.
generalized) 400–600 mg hepatotoxicity, metabolite (MHD) Dec’d by CBZ,
q1–2 weeks up hypoNa Inducer: CYP3A4, UGT PHB, PHT,
to 800 mg QD 1A1 (weak) PRM. Toxicity
(recommended), Inhibitor: CYP2C19 inc’d by CBZ,
or max dose (mod) OXC. Dec’s OCPs
1600 mg QD Protein binding: <40% (weak), warfarin.
Dose adjust: STerat/CTerat
renal. Avoid in unknown, BF
329
liver failure unknown (in milk)
OTHER (E.G.,
MONITORING,
SIDE EFFECTS DRUG
SEIZURE (COMMON, RARE METABOLISM/ INTERACTIONS,
NAME TYPE MECHANISM PO DOSING IV DOSING BUT BAD) BINDING LEVEL INFO PHARMACOKINETICS PREGNANCY)
Ethosuximide, Absence T-type Ca+ Initial: 500 mg/ N/V, hyperactivity Metabolism: hepatic 40–100 mcg/ T ½: 50–60 hr Neuropathic pain
ESX (Zarontin) seizures channels day SJS, DRESS, Enzyme: CYP3A4 and mL Tpeak: 1–7 hr med. Dec’d by
Depresses motor Inc by 250 mg agranulocytosis, non-CYP Check levels PHT. Inc’d by
cortex q4–7 days. bone marrow Protein binding: 5% after 1–2 weeks VPA. STerat
Usual dose: suppression moderate, CTerat
20–40 mg/kg unknown, BF
(divided QD-TID) unknown (in milk)
Ezogabine Focal KCNQ Initial: 100 mg Retinal pigmentary Metabolism: hepatic, T ½: 7–11 hr, Not available in
[within the USA], seizures voltage-gated K+ TID. Inc by abnormalities non-CYP increased by 30% in the
retigabine channels (GABA) ≤150 mg resulting in vision Active metabolite elderly USA. Ophthalmic
[outside the qweekly up to loss (~30% of N-acetyl active Tpeak: 0.5–2 hr, exam (acuity,
USA], (Potiga, 400 mg TID (no patients), with black metabolite (NAMR) delayed by 0.75 hr fundoscopy, OCT)
Trobalt) added benefit box warning. Protein binding: 80% when given with at baseline and at
>900 mg/day) Urinary retention high-fat food 6-month intervals
Elderly: 50 mg Dec’d by CBZ,
TID, up to PHT
250 mg TID STerat/CTerat
Dose adjust: unknown, BF
renal and unknown
hepatic
Felbamate, FBM Focal Inhibits NMDA, Initial: 1200 Anorexia, N/V, Metabolism: 50% T ½: 20–23 hr, CBC and LFT at
(Felbatol) seizures, augments GABA mg/day divided constipation, URI hepatic, 50% renal increased by 9–15 hr baseline and
Lennox- (TID-QID). Aplastic anemia excretion without in renal impairment q1–2 months,
Gastaut Inc by 600 mg (black box warning), metabolism Tpeak: 2–6 hr even after FBM
syndrome q2 weeks up to liver failure (black Enzyme: CYP3A4 dc’ed. Dec’d by
3600 mg/day box warning) (major), CYP2E1 CBZ, PHB, PHT,
Dose adjust: Protein binding 25% PRM. Dec’s OCPs
renal (weak) STerat/
CTerat unknown,
BF unknown (in
milk)
Gabapentin, GBP Focal Voltage-gated Initial: 300 mg Peripheral edema. Excreted entirely in 2–20 mcg/mL, T ½: 5–7 hr, prolonged Neuropathic pain
(Neurontin) seizures (can Ca+ channel, TID. urine, with no though not well in renal impairment med. Dec’d by
worsen inhibits NT Max dose: metabolism established Tpeak: 2–4 hr (IR) 8 hr antacids (give
generalized) release (Na+, 2400 mg/day Protein binding: <5% (ER) GBP 2 hr after
Ca+) (divided TID) antacids). STerat
Dose adjust: low-moderate,
renal CTerat unknown,
BF unknown (in
330
milk)
OTHER (E.G.,
MONITORING,
SIDE EFFECTS DRUG
SEIZURE (COMMON, RARE METABOLISM/ INTERACTIONS,
NAME TYPE MECHANISM PO DOSING IV DOSING BUT BAD) BINDING LEVEL INFO PHARMACOKINETICS PREGNANCY)
Lacosamide, LCM Focal Na+ channels Initial: 100 BID Second line N/V Metabolism: 30% T ½: 13 hr Obtain EKG
(Vimpat) seizures as monotherapy, for sz > 5 PR prolongation, CYP450, 20% Tpeak: 1–4 hr before starting,
(may or 50 BID as min: bradycardia, non-CYP. Enzyme: and at
exacerbate adjunct. Load: hypotension CYP2C19 (genetic maintenance dose
seizures in Inc qweekly 200– polymorphisms may Dec’d by CBZ,
Lennox- 50–100 mg/ 400 mg affect conc), 2C9, 3A4 PHT, PHB. Toxicity
Gastaut day up to 400 (over 30–60 Protein binding: <15% inc’d by CBZ.
syndrome) mg/day. Dose min) STerat/Cterat
adjust: renal, 1:1 PO:IV unknown, BF
hepatic conversion unknown (in milk)
Lamotrigine, LTG Broad Na+ channels, Initial: 25 QD Nausea. Rash, SJS, Metabolism: hepatic 2.5–15 mcg/ T ½: 25–33 hr Mood stabilizer.
(Lamictal) spectrum inhibits release of (QOD if hepatotoxicity Auto-induction of mL, though not (15–70 hr if drug Dec’d by CBZ,
(can worsen glutamate (Ca+) concurrent VPA) metabolism via well established interactions) PHT, PRM, OCPs.
myoclonic Inc by 50 mg UGT-glucuronidation Tpeak: 1–5 hr (IR) Inc’d by
seizure) QD q2 weeks, Protein binding: 55% 4–11 hr (ER) VPA. Toxicity inc’d
up to 375 mg by VPA. Dec’s
QD (500 mg OCPs (weak)
QD if on STerat low/safest
inducer). ER QD, (inc with dose).
IR BID. Dose CTerat low/safest.
adjust: for BF safe (in milk).
concurrent VPA/ Level dec in
CBZ/PHT/PHB pregnancy
Levetiracetam, Broad Synaptic vesicle Initial: First line for Irritability, mood Metabolism: 24% 10–45, though T ½: 6–8 hr Dec’d by CBZ.
LEV (Keppra) spectrum protein 2A 500–2000 mg sz > 5 min: changes (can occur hydrolysis in the blood, the effect is not Tpeak: 1 hr (IR) STerat low/safest
binding (Ca+, BID. Inc by 60 mg/kg, outside initial titration >65% renally excreted level dependent 4 hr (ER) CTerat low/ safest
K+, GABA) 500–1000 mg or 1–3 g period), weight loss. without metabolism Tpeak extended if taken BF safe (in milk)
q14 days (faster (max 4.5 g) Bone marrow Protein binding: <10% with food Level dec in
as inpatient) up over 15 min. suppression, pregnancy,
to 4000 mg/ 1:1 PO:IV eosinophilia, SJS starting in the first
day. Dose conversion trimester
adjust: renal
331
OTHER (E.G.,
MONITORING,
SIDE EFFECTS DRUG
SEIZURE (COMMON, RARE METABOLISM/ INTERACTIONS,
NAME TYPE MECHANISM PO DOSING IV DOSING BUT BAD) BINDING LEVEL INFO PHARMACOKINETICS PREGNANCY)
Lorazepam, LZ Abortive for Benzodiazepine, For sz <5 Hypotension, Metabolism: hepatic, T ½: 12 hr (PO) Olanzapine IM +
(Ativan) prolonged or inc freq of min: 2 mg respiratory conjugated to 14 hr (IV) LZ IV inc’s risk of
cluster GABA receptor For sz >5 depression lorazepam glucuronide T ½ CNS: 2–3 hr CV/respiratory
seizures Cl- channel min: 0.1 (inactive) Tpeak: 2 hr (PO) depression. Inc’d
opening mg/kg up to Protein binding: 90% Onset: <10 min (IV) by VPA
4 mg per STerat/CTerat/BF
dose unknown (in milk).
Risk of neonatal
withdrawal
Midazolam, MZ Abortive for Benzodiazepine, For sz <5 Hypotension, Metabolism: hepatic T ½: 3 hr STerat/CTerat
prolonged or inc freq of min: can be respiratory Enzyme: CYP3A4 Tpeak: 0.5–1 h (IM) unknown, BF
cluster GABA receptor given IM at depression, sedation Inhibitor: CYP2C8, Onset: 15 min (IM) unknown (in milk)
seizures Cl- channel 0.2 mg/kg CYP2C9 (weak) T ½ prolonged in renal
opening up to 10 mg dysfunction
per dose
Oxcarbazepine, Focal seizure Na+ channels Initial: N/V, hypoNa (inc Metabolism: hepatic, Optimal MHD T ½: Dec’d by CBZ,
OXC (Trileptal) (can worsen (Ca+, K+), 300–600 mg responsiveness to non-CYP level 2–55 2 hr (OXC IR) PHT, PHB, VPA,
generalized) similar to CBZ QD. ADH) Prodrug for active mcg/ml (maybe 7–11hr (OXC ER) PRM. Inc’d by
Inc up to Rash, DRESS, SJS monohydroxy 8–35), though 9 hr (MHD IR) PER.
2400 mg daily (test Asians for metabolite (MHD) no clear 9–11 hr (MHD ER) Dec’s OCPs
(divided BID or HLA-B*1502, which 30% renally excreted evidence for Tpeak: 3–13 hr, (strong).
TID). Can load increases risk), as active MHD therapeutic median 4.5 hr (IR); 7 hr STerat moderate,
with 30 mg/kg. hypothyroidism Inducer: CYP3A4 importance of (ER) CTerat unknown,
May require Protein binding: 40% level Prolonged in renal BF unknown (OXC
higher doses as as MHD impairment and MHD in milk)
ER Level dec in
Dose adjust: pregnancy
severe renal
impairment
332
OTHER (E.G.,
MONITORING,
SIDE EFFECTS DRUG
SEIZURE (COMMON, RARE METABOLISM/ INTERACTIONS,
NAME TYPE MECHANISM PO DOSING IV DOSING BUT BAD) BINDING LEVEL INFO PHARMACOKINETICS PREGNANCY)
Perampanel, PER Broad AMPA antagonist Initial: 2 mg Nausea, weight Metabolism: hepatic Monitor free T ½: 105 hr Dec’d by CBZ,
(Fycompa) spectrum Loading doses of qnightly gain, hostility/ Enzyme: CYP3A4/5 (unbound) conc Tpeak: 0.5–2.5 hr PHT, OXC, PRM
12-24 mg have Inc by 2 mg aggression with (major), 1A2/2BG in renal or (delayed 1–3 hr with Dec’s OCPs
been used for qweekly, up to suicidal/homicidal (minor), hepatic food) (strong)
Status. Discuss 12 mg QD ideation, rash. Acute glucuronidation impairment Takes 2 weeks to reach STerat/CTerat
with pharmacist Dose adjust: psychosis, DRESS, Protein binding: 95% On 6 mg QD, steady state unknown. BF
before using a renal, hepatic, hypertriglyceridemia average peak unknown
loading dose concurrent PHT/ conc is 460
CBZ/OXC ng/mL
On 12 mg QD,
average peak
conc is 800
ng/mL
Phenobarbital, Focal seizure Barbiturate, inc Initial: Second line Hypotension, Metabolism: hepatic, 10–40 mcg/ T ½: ~79 hr (53–118 Inc’d by OXC,
PHB/PB (some GABA duration 50–100 mg for sz > 5 bradycardia, 25% excreted renally mL, 25–50 in hr) PHT, RUF,
efficacy for (AMPA, Na+, BID-TID min: sedation, respiratory Enzyme: CYP2C19 status Tpeak: 0.5–4 hr PO VPA. Dec’s
generalized) Ca+, depresses Loading dose = 15–20 mg/ depression (major; genetic epilepticus. Onset: >1 hr (PO) warfarin, NOACs,
sensory cortex) desired kg (100 SJS, polymorphisms may Check 1–2 hr 5 min (IV) corticosteroids
level – measured mg/min), thrombocytopenia, affect conc), 2E1/2C9 after load Peak CNS depression Dec’s OCPs
level x (0.5 x can give agranulocytosis, (minor) As output, after IV dose is (strong)
ideal body additional megaloblastic Inducer: CYP2A6, check level in >15 min STerat high,
weight) 5–10 mg/kg anemia CYP3A4, UGTA1 3–4 weeks CTerat high,
Dose adjust: 1:1 PO:IV (strong) BF unknown (in
renal, hepatic conversion Protein binding: 55% milk)
Level dec in
pregnancy
333
OTHER (E.G.,
MONITORING,
SIDE EFFECTS DRUG
SEIZURE (COMMON, RARE METABOLISM/ INTERACTIONS,
NAME TYPE MECHANISM PO DOSING IV DOSING BUT BAD) BINDING LEVEL INFO PHARMACOKINETICS PREGNANCY)
Phenytoin, PHT Focal seizure Na+ channels IR: 100 mg TID First line for Bradycardia, Metabolism: hepatic 10–20 mcg/ml, Michaelis-Menten Folic acid (0.5
(Dilantin) (some (Ca+) To inc, given sz > 5 min: hypotension (IV), Enzyme: CYP2C19 15–20 in status kinetics: first-order mg/day) may dec
efficacy for kinetics, Phenytoin gingival hypertrophy, (genetic polymorphisms epilepticus. kinetics at low conc, risk of gingival
generalized consider only 20 mg/kg body hair increase, may affect conc), 2C9, Check 2 hr but 0-order at hyperplasia. Ca
but can also additional load, (<50 mg/ folic acid depletion, 3A4 after dose therapeutic conc and vitamin D in
worsen with dose = min), can decreased bone Inducer: CYP3A4, PGP, See (enzymes saturated, chronic therapy.
generalized) (desired give density UGT1A1 (strong), “Pharmacology metabolism rate Monitor EKG and
level – measured additional Arrhythmia (IV), bone CYP1A2, 2B6 (weak) Tips for constant). Thus, small BP with IV
level) × (0.7 × 5–10 mg/ marrow suppression, Protein binding: 90% Commonly dose changes can yield formulation
weight in kg). kg, or hepatotoxicity, rash, Used AEDs” for big conc changes Inc’d by BRV,
Use adjusted fosphenytoin DRESS, SJS (test correction in T ½: 7–42 hr, dose CBZ, ESL, ESX,
body weight if 20 PE/kg Asians for patients with dependent FBM, OXC, RUF,
obese (150 mg/ HLA-B*1502, which low albumin Tpeak: 1.5–3 hr (IR) CBD. VPA dec’s
Otherwise, min) can increases risk). Purple As outpatient, 4–12 hr (ER) PHT protein
adjust qweekly give glove syndrome (IV) check level 2–3 Onset: 0.5–1 hr (IV) binding, may inc
up to 600 mg/ additional 5 weeks after the free level Dec’d by
day mg/kg first dose CBZ, PHB,
ER: Load with 1 Monitor EKG VGB. Altered by
g in 3 doses 2 and BP CZP
hr apart (400, 1:1 PO:IV Dec’s OCPs
300, 300). Then conversion (strong), warfarin,
100 mg TID (or NOACs,
300 mg QD), corticosteroids
adjust qweekly STerat moderate,
up to 200 mg CTerat low, BF
TID safe (in milk)
Pregabalin, PGB Focal seizure Voltage-gated Initial: 150 mg/ Peripheral edema, >95% renally excreted T ½: 6.3 hr Used for
(Lyrica) Ca+ channel, day (divided weight gain, visual without metabolism Tpeak: 0.7 hr fasting, 3 neuropathic pain
inhibits NT BID-TID) loss Protein binding: 0% hr with food (IR) STerat/CTerat
release Inc up to 600 8 hr (ER) unknown. BF
mg/day unknown (in milk)
Dose adjust:
renal
334
OTHER (E.G.,
MONITORING,
SIDE EFFECTS DRUG
SEIZURE (COMMON, RARE METABOLISM/ INTERACTIONS,
NAME TYPE MECHANISM PO DOSING IV DOSING BUT BAD) BINDING LEVEL INFO PHARMACOKINETICS PREGNANCY)
Primidone, PRM Broad Barbiturate, inc Initial: 100 mg N/V Metabolism: 75% Follow level T ½ (age dependent): Used for tremor
(Mysoline) spectrum GABA duration qnightly. After 3 Bone marrow hepatic, 25% renally (and PHB level) PRM 5–16 hr, PEMA Inc’d by FBM,
(AMPA, Na+, days, BID. After suppression, rash excreted in renal or 16–50 hr, PHB ~79 hr VPA. Dec’d by
Ca+, depresses 3 days, Metabolized to hepatic Tpeak: 0.5–9 hr PHT. Toxicity inc’d
sensory cortex) TID. Usual dose phenobarbital and impairment. by TPM. Dec’s
750–1500 mg PEMA (which enhances Goal 5–12 OCPs (strong),
(divided activity of mcg/ml (SI warfarin, NOACs,
TID-QID), max of phenobarbital) 23–55 corticosteroids
2 g/day Inducer: CYP3A4 micromole/L). Pregnancy risk
Dose adjust: (strong), CYP1A2, Toxicity rare for presumed similar
renal, hepatic 2B6, 2C9 (weak) level <10 (SI to PHB: STerat
Protein binding: 10% 46). Toxicity high, CTerat high,
>15 (SI >69). BF unknown (in
Two weeks for milk)
steady state
Rufinamide, RUF Broad Prolongs inactive Initial: 400–800 N/V, QT interval Metabolism, hepatic, T ½: 6–10 hr Dec’d by CBZ,
(Banzel) spectrum, state of Na+ mg/day shortening non-CYP. Renally Tpeak: 4–6 hr, PHB, PHT, PRM.
Lennox- channels (divided BID), Bone marrow excreted prolonged by food Inc’d by VPA
Gastaut <400 mg/day if suppression Inducer: CYP 3A4 Dec’s OCPs
syndrome concurrent VPA (weak) (weak)
Inc by Inhibitor: CYP2E1 STerat/CTerat
400–800 mg (weak) unknown. BF
per day q2 days Protein binding: 35% unknown
up to 3200 mg/
day
Avoid in liver
failure
Tiagabine, TGB Focal seizure Inhibits GABA Initial: 4–8 mg Nausea, infection, Metabolism: hepatic Trough of T ½: 7–9 hr STerat/CTerat
(Gabitril) (can worsen reuptake QD. accidental injury Enzyme: CYP3A4 50–250 nmol/L Tpeak (fasting): 45 min unknown. BF
generalized) Inc by 4 mg Edema, rash Protein binding: 95% has been unknown
qweekly, up to suggested, not
32–56 mg/day well established
(BID-QID)
335
OTHER (E.G.,
MONITORING,
SIDE EFFECTS DRUG
SEIZURE (COMMON, RARE METABOLISM/ INTERACTIONS,
NAME TYPE MECHANISM PO DOSING IV DOSING BUT BAD) BINDING LEVEL INFO PHARMACOKINETICS PREGNANCY)
Topiramate, Broad Multi-(Na+, Ca+, Initial: 25 mg Paresthesia, Metabolism: <30% Level of 5–20 T ½: IR 12–24 hr, Monitor HCO3.
TPM/TOP spectrum GABA, BID metabolism acidosis hepatic non-CYP, >65% correlates with Qudexy XR 56 hr, Migraine ppx, IIH
(Topamax, antagonist of Inc by 50 mg (inhibits carbonic renally excreted response for Trokendi XR 31 hr treatment. Dec’d
Qudexy, NMDA) qweekly up to anhydrase, causing without metabolism most patients Tpeak: IR 2 hr by CBZ, PHT.
Trokendi) 200 mg BID renal bicarb loss), Inducer: CYP3A4 Qudexy XR 20 hr, >200 mg/day,
(400 mg QD ER) anorexia, weight Inhibitor: CYP2C19 Trokendi XR 24 hr dec’s OCPs
Dose adjust: loss, diarrhea. Protein binding: 10% (weak). STerat
renal, hepatic Nephrolithiasis, moderate-high
glaucoma (e.g., oral cleft),
CTerat unknown,
BF unknown (in
milk). Small for
gestational age
risk. Level dec in
pregnancy
Valproic acid, Broad Multi–(GABA, Initial: 15 mg/ First line for Hyperammonemia Metabolism: hepatic Total level: T ½: 9-19 hr Migraine ppx and
valproate, VPA spectrum, Na+, Ca+) kg/day sz > 5 min: (30% of pts, treat Enzyme: multiple Goal 50–100 Tpeak: mood stabilizer.
(depakote, idiopathic (regular/ 20–40 mg/ with levocarnitine), CYP450 mcg/ml, 4 hr (IR) Monitor LFTS 1–2
depakene, genetic depakene and kg (200 hair loss, N/V, Inducer: CYP2A6 70–140 in 4-17 hr (ER) times per year.
divalproex) epilepsy delayed/ mg/min), weight gain. (weak/mod) status Divalproex is
syndromes depakote can give Thrombocytopenia, Inhibitor: CYP2C9 epilepticus. valproate +
BID-QID; ER additional hypothyroidism, (weak) Check >1 hr valproic acid,
QD). Inc 20 mg/kg PCOS, pancreatitis, Protein binding: 95% after loading dissociates to
qweekly by 1:1 PO:IV parkinsonism dose. Toxicity valproate in GI
5–10 mg/kg/ conversion 100–150. As tract. Dec’d by
day, up to 60 output, check CBZ, PHT, OCPs.
mg/kg/day trough level Inc’d by FBM,
When 1–2 weeks, PRM. Toxicity
converting goal 50–125 inc’d by
depakote to ER: mcg/ml. Free TPM. Inc’s
inc dose by level goal: warfarin (dec’s
8–20% 5–15 mcg/ml protein binding).
Dose adjust/ STerat high,
caution: hepatic CTerat high, BF ok
if fetus already
exposed in utero
(in milk)
336
OTHER (E.G.,
MONITORING,
SIDE EFFECTS DRUG
SEIZURE (COMMON, RARE METABOLISM/ INTERACTIONS,
NAME TYPE MECHANISM PO DOSING IV DOSING BUT BAD) BINDING LEVEL INFO PHARMACOKINETICS PREGNANCY)
Vigabatrin, Focal seizure Irreversibly Initial: 500 mg N/V/D, visual loss >90% renally excreted T ½: 10.5 hr STerat/CTerat
VGB/VBT (Sabril) (can worsen inhibits GABA-T BID (black box warning), without metabolism Tpeak: 1 hr (2 hr with unknown. BF
generalized) enzyme, Inc by 500 mg blurred vision Protein binding: 0% food) unknown (in milk)
increasing GABA qweekly up to White matter
1500 mg BID changes, rash, URI
Dose adjust:
renal
Zonisamide, ZNS Broad Na+, Ca+ Initial: 100–200 Anorexia Metabolism: hepatic T ½: 63 hr Dec’d by PHT,
(Zonegran) spectrum, mg/day (QD or Metabolic acidosis, Enzyme: CYP3A4 Tpeak: 2–6 hr PHB
myoclonic BID) nephrolithiasis Protein binding: 40% STerat low, CTerat
epilepsy Inc up to 600 (carbonic anhydrase unknown, BF
mg/day. No inc inhibitor), decreased unknown (in milk).
response >400 sweating Small for
mg/day, with gestational age
inc adverse risk
effects >300
mg/day
Dose adjust:
renal, hepatic
(slow titration)
337
DRUG-DRUG INTERACTIONS COMMON
IN NEUROLOGY PATIENTS
Stephanie Seto, Amanda Rivera, and Megan E. Barra
DEFINITIONS [1]
• Substrate: A substrate is a drug that binds to a specific enzyme and metabolized by
that enzyme. Substrate metabolism may be affected by enzyme inhibitors or
enzyme inducers.
• Inducer: Compounds that either increase the production of the enzyme or increase
the activity of the enzyme, which results in increased metabolism and therefore
lower concentrations of substrate. The time course of induction is dependent on
drug half-life and time required to upregulate metabolizing enzymes and may take
several weeks to see effect. Enzyme induction may persist for several weeks after
inducer discontinuation and may result in decreased substrate efficacy.
• Inhibitor: Compounds that inhibit the activity of the enzyme, which results in
decreased metabolism and therefore higher concentrations of substrate. The time
course of inhibition dependent on drug half-life, but usually see the effects of
enzyme inhibitors on affected agents within 24–48 hours. Enzyme inhibition will
resolve several days after inhibitor discontinuation and increased the risk for
substrate toxicity.
340
CYP EXAMPLE OF SUBSTRATES [3] INDUCERS INHIBITORS
2C19 Neurologic Citalopram, phenytoin, Carbamazepine, Esomeprazole,
primidone phenobarbital, felbamate,
Analgesics Methadone phenytoin, rifampin fluconazole,
Anti-thrombotic Clopidogrel fluoxetine, isoniazid,
Anti-infective Voriconazole modafinil,
omeprazole,
voriconazole
2D6 Neurologic Aripiprazole, Amiodarone,
clozapine, donepezil, bupropion,
duloxetine, haloperidol, duloxetine,
risperidone, tricyclic fluoxetine,
antidepressants methadone,
Analgesics Codeine, hydrocodone, paroxetine,
meperidine, sertraline
oxycodone, tramadol
Cardiovascular Carvedilol, metoprolol,
propranolol
PgP Anti-thrombotic Apixaban, dabigatran, Carbamazepine, Amiodarone,
efflux rivaroxaban phenytoin, rifampin erythromycin,
Cardiovascular Digoxin verapamil, ritonavir
Note: This is not an all-inclusive list of drug-drug interactions, but an overview of strong, moder-
ate, or notable interactions with commonly used medications. Please use clinical judgment
before the concomitant use of other medications not listed in the table above.
a
Carbamazepine is an autoinducer; therefore, over time, carbamazepine will induce its own
metabolism resulting in lower serum concentrations of the drug
341
• Valproate-Phenytoin [2]
°V alproate displaces phenytoin from plasma protein binding sites, significantly
increasing free (active) phenytoin. Total phenytoin concentration may remain the
same or decrease. Exercise extreme caution when interpreting total phenytoin
levels on patients receiving both valproate and phenytoin.
° Bottom line: Avoid combination when possible. If combination therapy is
used, recommend monitoring free phenytoin levels.
• Phenytoin, Phenobarbital, Carbamazepine, and Anticoagulation
° DOACS: Phenytoin, phenobarbital, and carbamazepine significantly induces the
metabolism of DOACs (apixaban, dabigatran, rivaroxaban, edoxaban) and may
result in therapeutic failure. Avoid the combination of phenytoin, phenobarbital,
or carbamazepine with DOAC anticoagulants [4].
° Warfarin: Many AEDs impact warfarin; closely monitor INR during initiation and
dose changes of AED and/or warfarin.
REFERENCES
1. Spoelhof B, Farrokh S, Rivera-Lara L. Drug interactions in neurocritical care. Neurocrit Care.
2017;27(2):287–96.
2. Brodie MJ, Mintzer S, Pack AM, et al. Enzyme induction with antiepileptic drugs: cause for con-
cern? Epilepsia. 2013;54(1):11–37.
3. CYP450 drug interactions. Pharmacists Lett 2006.
4. Galgani A, Palleria C, Iannone LF, et al. Pharmacokinetic interactions of clinical interest between
direct oral anticoagulants and antiepileptic drugs. Front Neurol. 2018;9:1067.
5. Mori H, Takahashi K, Mizutani T, et al. Interaction between valproic acid and carbapenem antibi-
otics. Drug Metab Rev. 2007;39(4):647–57.
6. Al-Quteimat LA. Valproate interaction with carbapenems: review and recommendations. Hosp
Pharm. 2020;55(3):182–8.
342
MYASTHENIA GRAVIS: MEDICATIONS TO AVOID
Megan E. Barra and John Y. Rhee
MEDICATION
CLASS EXAMPLES COMMENTS
Antimicrobials [3]
Aminoglycosides Amikacin, gentamicin, Avoid use, unless no alternative therapy
tobramycin available. Impairs neuromuscular transmission.
Colistin/ Colistin, colistimethate Use cautiously and only if no alternative
polymyxin B sodium, polymyxin B treatment available. Impairs neuromuscular
transmission.
Fluroquinolones Ciprofloxacin, Avoid use, if no alternative available use very
levofloxacin, moxifloxacin cautiously. Black box warning for use in
MG. Disrupts neuromuscular transmission.
Lincosamides Lincomycin Avoid use. Pre- and post-synaptic effects on
neuromuscular junction.
Macrolides Azithromycin, Avoid use, if no alternative available use very
clarithromycin, cautiously. Telithromycin has black box warning
erythromycin, telithromycin for use in MG. Inhibits neuromuscular
transmission.
Penicillamine Penicillamine, Avoid use. Induces autoimmune myasthenia
penicillamine (D-) gravis and reported to occur in 1–7% of all
patients on penicillamine.
Antimalarial Chloroquine, Avoid use if possible, case reports of impaired
hydroxychloroquine [4] neuromuscular transmission with
hydroxychloroquine and chloroquine
utilization.
344
MEDICATION
CLASS EXAMPLES COMMENTS
Neuromuscular Depolarizing Relative resistance and prolonged duration of
Blocking agents neuromuscular blocking action due to decreased Ach receptors. Often
agents (e.g., higher doses are required (up to 2× normal
succinylcholine) dose). Inhibition of hydrolysis of succinylcholine
in patients on anti-cholinesterase inhibitors (e.g.
pyridostigmine) at baseline further prolongs
duration of action.
Nondepolarizing Increased sensitivity and prolonged duration of
Neuromuscular blocking action. Often significantly lower doses are
agents (e.g., required (50% of normal dose). May be
Cisatracurium, preferred neuromuscular blocker over
rocuronium, vecuronium) succinylcholine.
Steroids Hydrocortisone, May cause transient worsening of myasthenia
(Intravenous) dexamethasone, gravis within first 2 weeks of use. Initiation of
methylprednisolone, steroids for treatment of myasthenia gravis or
triamcinolone other indications should be closely monitored.
Many different agents have, in rare instances, been associated with worsening of
myasthenia gravis or development of new-onset myasthenia gravis in case
reports or in experimental studies exhibiting impaired neuromuscular
transmission. Clinical significance of these interactions is less clear compared to
those listed above and may include:
Cardiac agents Calcium channel blockers To mitigate risk with these agents: Use lowest
(e.g. verapamil), statins dose necessary and observe for worsening of
[6] (e.g. atorvastatin, symptoms
rosuvastatin, simvastatin) • Worsening respiratory symptoms with NIF
Antimicrobials Bactrim and VC either trending down. High concern
(sulfamethoxazole/ if NIF < 20 cm H2O and/or VC <1 L. Can
trimethoprim), also check with weakening neck flexion,
clindamycin, doxycycline, inability to count to >20 with one breath,
nitrofurantoin, ampicillin rapid worsening of weakness of baseline
Antiepileptic Carbamazepine, myasthenia weakness.
drugs [9] ethosuximide, gabapentin,
phenobarbital, phenytoin
CNS agents Amitriptyline,
dexamphetamine,
imipramine, haloperidol,
lithium
Misc [10, 11] Riluzole, glatiramer
Avoid use
Consider other agents
Case reports, unclear significance
345
REFERENCES
1. Mehrizi M, Fontem RF, Gearhart TR, Pascuzzi RM. Medications and myasthenia gravis (a ref-
erence for health care professionals). Myasthenia Gravis Foundation of America; 2015. http://
www.myasthenia.org/portals/0/draft_medications_and_myasthenia_gravis_for_MGFA_web-
site_8%2010%2012.pdf.
2. Myasthenia Gravis Foundation of America. Cautionary drugs. New York: Myasthenia Gravis
Foundation of America, Inc; 2019.
3. Jones S, Sorbello A, Boucher R. Fluoroquinolone-associated myasthenia gravis exacerbation:
evaluation of postmarketing reports from the US FDA adverse event reporting system and a
literature review. Drug Saf. 2011;34(10):839–47.
4. Jallouli M, Saadoun D, Eymard B, et al. The association of systemic lupus erythematosus and
myasthenia gravis: a series of 17 cases, with a special focus on hydroxychloroquine use and a
review of the literature. J Neurol. 2012;259(7):1290–7.
5. Jonkers I, Swerup C, Pirskanen R, et al. Acute effects of intravenous injection of beta-
adrenoreceptor- and calcium channel at antagonists and agonists in myasthenia gravis. Muscle
Nerve. 1996;19(8):959–65.
6. de Sousa E, Howard J. More evidence for the association between statins and myasthenia gra-
vis. Muscle Nerve. 2008;38(3):1085–6.
7. Safa H, Johnson DH, Trinh VA, et al. Immune checkpoint inhibitor related myasthenia gravis: sin-
gle center experience and systematic review of the literature. J Immunother Cancer. 2019;7:319.
8. Lee SC, Ho ST. Acute effects of verapamil on neuromuscular transmission in patients with myas-
thenia gravis. Proc Natl Sci Counc Repub China B. 1987;11(3):307–12.
9. So EL, Penry JK. Adverse effects of phenytoin on peripheral nerves and neuromuscular junction:
a review. Epilepsia. 1981;22(4):467–73.
10. Restivo DA, Bianconi C, Ravenni R, et al. ALS and myasthenia: an unusual association in a
patient treated with riluzole. Muscle Nerve. 2000;23(2):294–5.
11. Frese A, Bethke F, Lüdemann P, et al. Development of myasthenia gravis in a patient with mul-
tiple sclerosis during treatment with glatiramer acetate. J Neurol. 2000;247(9):713.
346
PARKINSON’S DISEASE: MEDICATIONS TO AVOID
Amanda Rivera and Megan E. Barra
348
REFERENCES
1. DeMaagd G, Philips A. Parkinson’s disease and its management: part 3 Nondopaminergic and
nonpharmacologic treatment options. PT. 2015;40(10):668–79.
2. Grissinger M. Delayed administration and contraindicated drugs place hospitalized Parkinson’s
patients at risk. PT. 2017;43(1):10–2.
3. Parkinson’s Disease Update Quality Measurement Development Work Group. Am. Acad. Neurol.
Minneapolis, MN; 2015:1–56.
4. Rebecca Gilbert. Medications to be avoided or used with caution in Parkinson’s disease.
American Parkinson Disease Association. New york; 2018: p. 1–10.
349
NIH Stroke Scale [1, 2]
352
353
NIH Stroke Scale [1, 2]
354
355
NIH Stroke Scale [1, 2]
356
357
REFERENCES
1. Brott T, et al. Measurements of acute cerebral infarction: a clinical examination scale. Stroke. 1989;20(7):864–70.
2. Lyden P, Brott T, Tilley B, Welch KM, Mascha EJ, Levine S, et al. Improved reliability of the NIH Stroke Scale
using video training. NINDS TPA Stroke Study Group. Stroke. 1994;25:2220–6.
358
Index
361
Intracranial hemorrhage (ICH) in infectious/inflammatory/neoplastic
anticoagulation management, 225–227 conditions, 42–43
landmark trials, 211–213 sequences, 37–38
venous thromboembolism prophylaxis, 318 in stroke, 39–40
Intracranial pressure (ICP), 50 Malignant middle cerebral artery infarction, 199–203
invasive ICP monitoring, 190–191 Maximum intensity projection (MIP), 36
management of, 193–196 Mean arterial blood pressure (MAP), 187–189
theory and formulas central to neurocritical MAP, see Man arterial blood pressure (MAP)
care, 187–189 Mechanical thrombectomy (MT), 66, 109–112
waveform interpretation, 191–192 Medial longitudinal fasciculus (MLF) injury, 15
Intracranial stenosis, 114 Meningitis
Intraparenchymal fiberoptic sensor, 191 infectious
Intraparenchymal hemorrhage CNS penetration, 149–150
admission checklist, 207 CSF findings, 148
early management, 207, 210 signs, 145
labs, 207 treatment, 148–149
non-traumatic, 205–206 workup, 146–147
ongoing management, 207 non-infectious, 151–153
scoring systems, 208–209 seizure prophylaxis, 316
Intravenous fluid compositions, 325–326 Mental status examination, 10
Invasive neuro-monitors, 190 Microbleeds, 226
Ischemic stroke, 50 Middle meningeal artery (MMA) embolization, 248
admission checklist, 75–77, 90 Miosis, 13
anticoagulation, 99–103 Moya-moya disease
aspirin vs. anticoagulation, 92 definition, 292
CAS, 95–97 major intraoperative complications, 293
CEA, 95–97 postoperative complications, 294
extracranial vs. intracranial, 91 procedure, 293
history, 89 vascular craniotomy-specific postoperative
pathophysiology, 90 orders, 295
seizure prophylaxis, 315 MRI, see Magnetic resonance imaging (MRI)
signs and symptoms, 89 Multiple sclerosis (MS), 175, 177
symptomatic definition, 95 McDonald criteria, 175, 176
venous thromboembolism prophylaxis, 318 Myasthenia gravis, 173, 270–271
medications class, 343–345
Mydriasis, 13
L
Lacunar syndrome, 27
Large vessel occlusion (LVO) N
anterior circulation occlusion, 65–66 Neurogenic shock, 277
outcomes, 64 NeuroICU
posterior circulation occlusion, 66 hypernatremia, 307–309
Lateralized periodic discharges (LPDs), 54, 265 hyponatremia, 311–312
Lateralized rhythmic delta activity (LRDA), 265 intravenous fluid compositions, 325–326
L’Hermitte sign, 175 nutrition, 303–305
Lindegaard Ratio (LR), 49 pressors and inotropes, 313–314
Linnoila and Pittock and the Mayo Clinic seizure prophylaxis, 315–316
Laboratories antibody matrix, 163–164 venous thromboembolism prophylaxis, 317–319
Lumbar drains (LDs), 197 Neurologic complications, 254
LVO, see Large vessel occlusion (LVO) Neuromuscular disease, 319
Neuromyelitis optica (NMO), 177
spectrum disorders, 155
M Neuroprognosis
Magnetic resonance imaging (MRI) history, 255
contrast enhancement, 45 monitoring for, 254
diffusion restriction patterns, 44 perspective in, 255
in hemorrhage, 41 Neuroprotection, 254
362
Neurovascular pathologies, 292–297 neurological exam, 285, 287
New onset weakness wakes up from surgery, 285
differential diagnosis, 167 Potential injury, anatomical locations of, 167
evaluation, 168 Pre-rounding patient
Guillain-Barré, selected variants of, 173 on ICU patients, 5
selected screening evaluation, 169–172 neuro floor patients presentation, 3–4
upper and lower motor neuron findings, 168 NeuroICU patients presentation, 6–7
Non-contrasted head CTs (NCHCT) on neurology patients, 3
acute hemorrhage, 32 PRES, see Posterior reversible leukoencephalopathy
herniation syndrome, 33 syndrome (PRES)
hydrocephalus assessment, 33 Pressors and inotropes, in NeuroICU, 313–314
indications, 29 Pulsatility index (PI), 49
neuroanatomy, 30–31
in stroke, 34–36
Nonconvulsive status epilepticus (NCSE), 259 R
Non-traumatic intraparenchymal Radioimmunoassay (RIA), 163
hemorrhage, 205–206 RCVS, see Reversible cerebral vasoconstriction
Normothermia, 253, 255 syndrome (RCVS)
Nutrition, in NeuroICU, 303–305 Refractory status epilepticus (RSE), 259
Respiratory failure, with Guillain-Barré
syndrome, 269
O Respiratory insufficiency, 277
Oculomotor (CN III) nerve palsy, 14 Reversal of selected antithrombotics, 215–219
One-and-a-half syndrome, 16 Reversible cerebral vasoconstriction
Optic neuritis, 155 syndrome (RCVS)
Oral anticoagulants, 215–218 clinical features, 123
imaging characteristics, 124
management principles, 125
P medications, 123
Paraneoplastic antibody panel, 163 Reversing direct factor Xa-inhibitor-related
Parenteral anticoagulants, 218 hemorrhages, 221–223
Parkinson’s disease, medications class, 347–348 Rhombencephalitis, 159
Perfusion imaging Rhythmic and periodic patterns, 264
ischemic core vs. penumbra, 72–73
techniques, 71
Perimesencephalic non-aneurysm SAH, 229 S
Peripheral weakness, etiology of, 169 Seattle Severe traumatic Brain Injury Consensus
Pressure reactivity index (PRx), 189 Conference (SIBICC) algorithm, 193
PFO closure, 114–115 Seizure, 137–138
Polyneuropathy, 269 Seizure prophylaxis, in NeuroICU, 315–316
Posterior reversible leukoencephalopathy Severe traumatic brain injury (sTBI), 193
syndrome (PRES) Spinal cord injury
clinical features, 123 ICU management of
imaging characteristics, 124 admission checklist, 277–278
management principles, 125 classic syndromes, 279
medications, 123 prognostication, 280
seizure prophylaxis, 316 steroids and spinal injury, 278
Postoperative craniotomy subacute to late complications, 280
complications, 285–286 venous thromboembolism prophylaxis, 319
diffuse pneumocephalus, 285, 287 Spinal shock, 277
seizure prophylaxis, 315 Sporadic epileptiform discharges, 264
tension pneumocephalus, 285, 287 Status epilepticus, 259–262
Postoperative management adjunctive workup, 261–262
cerebrovascular patients, 291–297 definitions, 259
craniotomy patient medications, 260–261
checklist for admission, 284 treatment preparation, at onset of
general admission orders, 284 seizure, 259–260
363
Stroke middle cerebral artery, 48
anterior circulation, 21–24 monitoring, 49
anticoagulation, 105–107 posterior cerebral artery, 48
anti-platelets, 105 principles, 47
arterial hypercoagulable state, 79–80 SAH, 49
computed tomography, 34–36 Transient ischemic attack (TIA), 112–113
etiology, 87 Transverse myelitis, 168
lacunar syndrome, 27 Traumatic brain injury (TBI)
magnetic resonance imaging, 39–40 dexamethasone, 248
posterior circulation, 25–27 emergent management, 245–246
thromboembolic disease, 82–83 focal lesions, 248
vasculopathy, 84–86 framework for, 245
venous hypercoagulable state, 81 Glasgow Coma Scale, 245
Subarachnoid hemorrhage (SAH) ICU management, principles of, 247
aneurysmal SAH middle meningeal artery embolization, 248
admission and early management, 231–232 monitor ICP, 246
daily management principles, 235–239 seizure prophylaxis, 315
application in, 49 surgical management, 248
differential, 229–230 trials, 251–252
notable trials, 241–243 venous thromboembolism prophylaxis, 318
scoring systems, 233 Trochlear (CNIV) nerve palsy, 14
Subjective Data, Objective Data, Assessment, Plan
(SOAP) style, 3–4
Sub-occipital/retrosigmoid/translabyrinthine U
craniotomies, 288 Uhthoff phenomenon, 175
Superrefractory status epilepticus (SRSE), 259 Urinary retention, 277
Syndrome of inappropriate anti-diuretic hormone
(SIADH), 311–312
V
Vascular lesions, seizure prophylaxis, 315
T Vasculopathy, 84–86
TBI, see Traumatic brain injury (TBI) Venous sinus thrombosis (VST)
TCDS, see Transcranial dopplers TCDS acute treatment, 119
Temporal lobe encephalitis anatomy, 117
with HSV limbic encephalitis, 159 chronic treatment, 119–120
with post-HSV anti-NMDA-R encephalitis, 157 neuroimaging findings, 118
Tenecteplaste (TNK), 62–64 risk factors, 117–118
Tension pneumocephalus, 287 symptoms, 118
Thromboembolic disease, 82–83 workup, 119
TIA, see Transient ischemic attack (TIA) Venous thromboembolism prophylaxis, in NeuroICU
Tissue factor pathway inhibitor (TFPI), 221 indications, 318–319
Tissue plasminogen activator (tPA) pharmacologic agents, 317
criteria for, 62 VITAMIN Mnemonic, 169
mechanical thrombectomy, 66, 109–112 VST, see Venous sinus thrombosis (VST)
TNK screening, 62–64
tPA, see Tissue plasminogen activator (tPA)
Transcranial dopplers (TCDS) W
application of, 47, 50 Western blot (WB), 163
364