Вы находитесь на странице: 1из 14

68  Stroke in Children

Meredith R. Golomb, José Biller

CHAPTER OUTLINE rate of arterial ischemic stroke around 1 in 4000 neonates


(Lynch and Nelson, 2001). Laugesaar et al. (2007) looked at
all prospectively and retrospectively diagnosed perinatal
STROKE AND THE DEVELOPING CEREBROVASCULAR stroke in Estonia and found a rate of 1 in 1587 live births.
SYSTEM DeVeber and colleagues (2001) found a rate of 0.67 cases of
EPIDEMIOLOGY cerebral venous thrombosis (CVT) per 100,000 children per
Full-Term and Near-Term Neonates year, with neonates making up 43% of cases; rates were not
The General Population of Children described in relation to the number of term births.
Chapter 111 discusses cerebral vascular injury in the prema-
High-Risk Subgroups
ture neonate.
PRESENTATIONS
ETIOLOGY The General Population of Children
Cardiac
Estimates of the incidence of all pediatric stroke in the United
Hematological
States and France have ranged from 2.6 to 13 cases per 100,000
Trauma and Vascular Compression children per year (Giroud et al., 1995), with some variation
Infection among studies on the inclusion of neonates, traumatic strokes,
Vascular Malformations/Vasculopathy/Migraine and meningitis, and whether to use 16 or 18 as the cutoff age
Drugs/Toxins for pediatric stroke. A review of 13 years of data in New Jersey
Metabolic found a stroke incidence of 2.24 per 100,000 per year, with
Gender and Ethnicity roughly equal rates of ischemic and hemorrhagic stroke
Genetic (Gandhi et al., 2012),
DIFFERENTIAL DIAGNOSIS
EVALUATION High-Risk Subgroups
History and Physical Certain subgroups of children are at high risk for stroke, with
Imaging Studies rates approaching or surpassing those in older adults. Medical
Coagulation Workup conditions may place children at risk for intracranial hemor-
Cardiac Evaluation rhage, ischemic stroke, or both.
Some types of central nervous system (CNS) vascular mal-
Other Studies
formations (e.g., arteriovenous malformations, telangiecta-
TREATMENT sias) are part of well-known neurocutaneous syndromes such
The Acute Period and Initiating Chronic Therapy as Sturge Weber, Osler–Weber–Rendu, Louis–Bar syndrome,
Additional Issues in Chronic Therapy Wyburn–Mason syndrome, and Klippel–Trenaunay syndrome.
Other Issues Vascular malformations may present with intracranial hemor-
rhage. A review of 22 cases of spontaneous intracranial hemor-
SUMMARY rhage seen over 10 years at two Swiss hospitals found that 55%
had arteriovenous malformation (Fig. 68.1), 18% had aneu-
rysm, 14% had cavernous malformation, and 14% had unclear
etiology (de Ribaupierre et al., 2008). Cavernous malforma-
tions and aneurysms may be genetic and can present in child-
STROKE AND THE DEVELOPING hood. Risk varies depending on the mutation involved (Denier
CEREBROVASCULAR SYSTEM et al., 2006). Brain aneurysms are rare in children and are
often dysplastic. The carotid bifurcation is the most common
Unlike adults, in most children with stroke, conditions such
site for pediatric intracranial aneurysms, followed by the pos-
as diabetes and hypertension make little contribution to the
terior circulation. Intracranial hemorrhage may lead to vaso­
etiology of stroke. Developmental, genetic, and environmen-
spasm and resultant ischemic stroke, but this is less common
tal factors are the major contributors to cerebrovascular injury
in children than in adults (Menkes et al., 2000).
in children.
Children with bleeding disorders are at high risk for intra­
cerebral hemorrhage. A study of intracranial hemorrhage in
EPIDEMIOLOGY Italians with hemophilia found the highest rate during the first
year of life, 24.4 per 1000, with a drop to 14.9 per 1000 in the
Full-Term and Near-Term Neonates second year of life (Zanon et al., 2012).
Neonates appear to be at higher risk for stroke than older Children with sickle cell anemia are at risk for ischemic
children. Asymptomatic subdural hemorrhage affects almost stroke because sickling red blood cells may lead to thrombosis
half of term neonates and can occur in infants delivered by or endothelial injury and in some patients are associated with
both vaginal and cesarean delivery (Rooks et al., 2008). Symp- moyamoya syndrome (Pegelow, 2001; Pegelow et al., 2002)
tomatic intracranial hemorrhage affects 1 in 100 full-term (Fig. 68.2). The rate of stroke in children with sickle cell
neonates (Gradnitzer et al., 2002). Most estimates place the anemia has dropped since the institution of transfusion

996
Stroke in Children 997

68

A B C D
Fig. 68.1  Axial and coronal T2W MRI and AP and lateral views of catheter cerebral angiography of a 17-year-old female with a left
thalamic arteriovenous malformation (Grade III Spetzler–Martin) fed from the PCA territories bilaterally.

A B C
Fig. 68.2  Magnetic resonance imaging (MRI) of a 10-year-old girl with sickle cell anemia. A–C, At age 5, she presented with a left hemi-
paresis. MRI then showed acute ischemia in the right middle cerebral artery (MCA) territory and chronic ischemic changes in the left cerebral
hemisphere. Magnetic resonance angiography (MRA) showed stenoses in both anterior circulations. These stenoses progressed over time, and
she was treated with pial synangiosis and burr holes. There was no clear progression of ischemic lesions. A, MRA demonstrates complete occlu-
sion of the left M1 segment of the MCA (solid arrow), and severe stenosis of the M1 segment of the right MCA (open arrow) and both A1 seg-
ments of the anterior cerebral arteries. There are multiple small vessels at the stenotic sites consistent with moyamoya. There appears to be an
anastomosis between the left superficial temporal branches and the distal left M1. B, C, Fast spin echo inversion recovery MRI demonstrates
multiple old infarcts in the bilateral frontal and parieto-occipital regions and centrum semiovale.

therapy (see the later section Treatment). In California, the infarcts from drops in perfusion pressure, and cerebral venous
incidence of first stroke in children with sickle cell anemia thrombosis (CVT) (Fig. 68.3). The rates of stroke in children
dropped from 0.88 per 100 person-years to 0.17 per 100 with complex congenital heart disease also vary among series,
person-years (Fullerton et al., 2004); however, approximately with some of the variation due to the severity of the malforma-
30% of children aged 5–15 years old have silent infarcts tion, the number of corrective surgeries required, anesthetic
(DeBaun et al., 2012). Children with sickle cell anemia may techniques during surgery, patient selection, and length of
also develop aneurysms and resultant intracranial hemor- follow-up. A large Canadian study looked at 5526 children
rhage, but this is more common in adults with sickle cell who underwent cardiac surgery and found a stroke rate of 5
anemia (Pegelow, 2001). per 1000 children (Domi et al., 2008). Mayer and associates
Children treated with mechanical circulatory support are (2002) found evidence of cerebrovascular complications in 5
at increased risk for both intracranial hemorrhage and embolic of 77 (6.5%) pediatric patients with heart transplants and an
ischemic stroke. The rates of infarction after extracorporeal average of 59.2 months of follow-up; 25 of the 77 patients
membrane oxygenation (ECMO) vary dramatically among (32.5%) died. The greatest risk for children with congenital
series, ranging from 0% to 26%; one study examined the heart disease occurs at the time of surgery or cardiac catheteri-
brains of 44 patients who died while on ECMO and found zation. One study found that 27% of children with stroke
evidence of focal ischemic infarct in 50% and intracranial associated with cardiac disease had recurrent stroke. Mechani-
hemorrhage in 52%. A recent large study of ECMO survivors cal heart valves, prothrombotic conditions, and infection at
found cerebral infarction in 7% and intracranial hemorrhage the time of first stroke all raised the risk of stroke recurrence
in 7% (Barrett et al., 2009). Stroke occurred in 7 of 17 treated (Rodan et al., 2012).
children (41%) at one center that started to use the Berlin Children with cancer are at risk for both intracranial hem-
Heart EXCOR ventricular assist device (Rockett et al., 2008); orrhage and ischemic infarction. Intracranial hemorrhage
however, the incidence of strokes with the EXCOR dropped occurs secondary to thrombocytopenia from bone marrow
with institutional experience (Byrnes et al., 2013). suppression or bleeding within the tumors. Children with
Children with complex congenital heart disease are at cancer may develop ischemic infarction or CVT due to
risk for cardioembolic stroke, thrombotic stroke, watershed leukostasis in the setting of leukemia; complications of
998 PART III  Neurological Diseases and Their Treatment

A B
Fig. 68.3  Magnetic resonance imaging (MRI) of a 3-year-old girl with a history of complex congenital heart disease. At age 10 months,
she had a procedure to repair a double inlet left ventricle and left transposition of the great arteries. She presented with seizures and dysconjugate
gaze 3 weeks later, and an acute right pontine infarct was diagnosed. Magnetic resonance angiography and catheter angiography demonstrated
narrowing of the distal basilar artery and the right superior cerebellar artery. At age 3, she presented with acute left hemiparesis and the following
imaging: diffusion-weighted MRI (A) and fast spin echo inversion recovery MRI (B) demonstrate multiple infarctions in the right temporal lobe,
inferior parietal region, and lentiform nucleus, consistent with cardioembolic stroke.

A B
Fig. 68.4  Magnetic resonance imaging (MRI) of an 8-year-old boy with leukemia who developed headaches and left hand numbness
during induction chemotherapy with L-asparaginase. A, Fast spin echo inversion recovery MRI demonstrates multifocal bilateral hemorrhagic
infarctions. B, Magnetic resonance venogram demonstrates irregularity of the superior sagittal sinus consistent with thrombosis (arrow).

chemotherapy such as L-asparaginase, which cause decreases abnormalities of the cervical spine, which increase risk for
in antithrombin, fibrinogen, and plasminogen; fungal or bac- vertebral artery dissection. Children with neurofibromatosis
terial meningitis leading to arteritis; vasculopathy secondary type 1 have higher-than-average rates of moyamoya and other
to radiation; or complications of intracranial surgery (Fig. occlusive vasculopathies that are sometimes but not always
68.4). Stroke affects approximately 1% of children with cancer, associated with radiation. Connective tissue disorders such as
most commonly children with leukemia or brain tumors Marfan and Ehlers–Danlos syndromes and pseudoxanthoma
(Noje et al., 2013). One institutional study found that 4% of elasticum may predispose to cervicocephalic arterial dissection
children treated with cranial and/or cervical radiation had or aneurysmal dilatation. Metabolic syndromes that damage
stroke during the 10 years after radiation (Mueller et al., 2013). the endothelium (e.g., homocystinuria, Fabry disease, familial
Children with certain syndromes are predisposed to stroke, hyperlipidemia) may predispose to vascular damage and
sometimes for multiple reasons. Children with Down syn- thrombosis.
drome have higher-than-average rates of leukemia, which in
turn may lead to hemorrhagic stroke; moyamoya vasculopa-
thy, which may lead to ischemic or hemorrhagic stroke;
PRESENTATIONS
complex congenital heart disease, which may lead to cardio­ Seizures are the most common manifestation of intraventricu-
embolic stroke; and atlantoaxial instability and other lar hemorrhages (IVH), arterial ischemic strokes, and CVT in
Stroke in Children 999

term neonates. Seizures are a presenting sign for 65% of term widespread; Cornelissen and colleagues (1996) found that
neonates with IVH, at least 80% of term neonates with arterial administering vitamin K lowered the incidence of vitamin K 68
ischemic stroke (Volpe, 2001), and over 50% of neonates with deficiency bleeding from 7 to 1.1 per 100,000 births per year.
CVT (Fitzgerald et al., 2006). Other presenting signs include However, late-onset vitamin K deficiency bleeding can occur
apnea, irritability, jitteriness, lethargy, and bulging fontanel. in children with undiagnosed cholestatic jaundice who
The immature central nervous system may not demonstrate received oral vitamin K and are exclusively breastfed, because
focal signs, and hemiparesis may not be apparent until a child they cannot absorb the vitamin, and breast milk is low in
is older than 6 months of age. vitamin K (Ijland et al., 2008). Schulte and colleagues (2014)
Children older than 6 months of age may present with reported a rise in late-presentation vitamin K deficiency bleed-
seizures or focal signs similar to those seen in adult stroke, ing in Tennessee because parents were refusing vitamin K,
with hemiparesis, ataxia, or aphasia. Children younger than 1 believing it was unnecessary or that the vitamin K injection
year of age are more likely to present with seizures and altered included “toxins.” Accidental ingestion or overdose of warfa-
mental status, whereas children older than 1 year of age are rin has the same effect as a vitamin K deficiency and may occur
more likely to present with focal motor signs (Zimmer et al., when a young child finds an older family member’s medica-
2007). Although severe headaches such as those in intracranial tions. The most common congenital coagulation factor defi-
hemorrhages often prompt parents to seek medical attention ciencies are deficiencies of coagulation factor VIII (hemophilia
immediately, most children arrive at the emergency room A), coagulation factor IX (hemophilia B), and von Willebrand
more than 6 hours after the event (Gabis et al., 2002). Parents factor. Intracranial hemorrhage is the most common cause of
may not detect focal motor weakness in a young child who death from bleeding in patients with hemophilia; the site of
has not yet started to walk or aphasia in a young child who is intracranial hemorrhage can be epidural, subdural, or intrapa-
just starting to speak. Parents may interpret the sudden onset renchymal. Spinal cord compression may result from spinal
of focal neurological signs as behavioral rather than neuro- hematomas.
logical. It can be easier to detect focal neurological signs and A deficiency or imbalance of factors involved in regulating
symptoms in older children, but these are also often missed coagulation may place the child at risk for thrombosis. Pedi-
in the first minutes to hours, possibly because many parents atric ischemic stroke has been associated with iron-deficiency
and children do not realize children can have strokes. Some anemia (Maguire et al., 2007), deficiencies of protein C,
children may have no symptoms or only gradual onset of protein S, and antithrombin; activated protein C resistance
developmental delay. While silent strokes are recognizable in due to the factor V Leiden mutation; the prothrombin gene
the sickle cell population, and several studies have screened 20210A mutation; the methylene tetrahydrofolate reductase
for them, the rates of silent infarction in other cerebrovascular (MTHFR) gene variants; the ATG haplotype of the protein Z
disorders are unclear because radiological investigations are gene (Nowak-Göttl et al., 2009); elevated lipoprotein (a);
lacking in the absence of clinical manifestations. elevated antiphospholipid antibodies (Kenet et al., 2010) and
lupus anticoagulant; elevated factor VIII levels; and low plas-
minogen or high fibrinogen. The importance of the plasmino-
ETIOLOGY gen activator inhibitor promoter polymorphism (PAI-1) in
Cardiac childhood stroke is controversial. Temporary hematological
abnormalities and resultant thrombosis may result from inter-
Complex congenital heart disease may lead to thrombosis current illness; for example, idiopathic nephrotic syndrome is
and ischemic stroke through several mechanisms. Abnormal associated with decreased antithrombin levels and CVT and/
cardiac anatomy or associated cardiac arrhythmias lead to or arterial thromboembolic events (Fluss et al., 2006).
abnormal flow and may predispose to the formation of int- Abnormalities of blood cells or blood cell concentration
racardiac thrombi. Septal defects may lead to right-to-left may place the child at risk for hemorrhagic or ischemic stroke.
shunts that allow venous thrombi to cross to the arterial side Stroke occurs in children with β thalassemia intermedia (βTI)
and cause cerebral infarction. Surgery and cardiac catheteriza- and children with paroxysmal nocturnal hemoglobinuria.
tion can disrupt the endothelium and lead to thrombosis. Low platelet count due to autoimmune thrombocytopenia or
Cardiac surgery itself may lead to a temporary prothrombotic bone marrow suppression leads to hemorrhage. Anything that
state (Petaja et al., 1996). An abnormal heart valve can serve increases blood viscosity (e.g., sickled cells, polycythemia,
as a nidus for bacterial or fungal vegetations that may cause chronic hypoxia) may predispose a child to arterial or venous
cardioembolic stroke. Chronic hypoxemia in severe cases of infarct. Dehydration is associated with arterial strokes and
congenital heart disease may lead to polycythemia, and the CVT, possibly because it increases viscosity. Anemia has also
increased blood viscosity may promote thrombosis. Aortic been associated with arterial ischemic infarction and CVT,
coarctation may be complicated by infective endarteritis or possibly due to alterations in hemodynamics or imbalances
ischemic or hemorrhagic strokes, including brain aneurysms. in thrombotic pathways.
Based on autopsy findings, a patent foramen ovale (PFO) is Several authors have noted the presence of multiple pro-
found incidentally in 20%–35% of the general population. thrombotic abnormalities in some children with ischemic
Although a PFO is more common in patients with cryptogenic stroke (Kenet et al., 2010). The combination of MTHFR and
stroke than in the general population, the relationship between endothelial nitric oxide synthase gene variants may raise
a PFO and ischemic strokes without a clearly identifiable stroke risk more than MTHFR alone (Djordjevic et al., 2009).
source in both children and adults remains controversial. Children with congenital heart disease or leukemia may be
at higher risk for developing thrombotic complications
during hospitalization if they also have a prothrombotic
Hematological abnormality.
Any hematological disorder that disrupts coagulation can
place a child at risk for hemorrhagic stroke. Newborns have
lower levels of coagulation factors and have a drop in the
Trauma and Vascular Compression
vitamin K-dependent factors in the first days of life. Bleeding Trauma is a risk factor for both ischemic and hemorrhagic
due to vitamin K deficiency was more common before stroke. Trauma can injure vessels directly, leading to hemor-
intramuscular or oral administration to neonates became rhage from torn vessels, thrombosis in damaged intima, or
1000 PART III  Neurological Diseases and Their Treatment

traumatic pseudoaneurysm. Subdural hemorrhages, subarach- Vascular Malformations/Vasculopathy/Migraine


noid hemorrhages, and ischemic infarctions occur in head
injury. Ascertaining the cause of the trauma is important; at As discussed previously, vascular malformations may present
one pediatric trauma center, the mortality rate for nonacciden- with intracerebral hemorrhage, and resulting vasospasm may
tal trauma was 9.7%, while the mortality rate for accidental lead to ischemic infarction. Arterial abnormalities such as
trauma was 2.2% (Roaten et al., 2006). Most subdural hemor- large-artery stenosis are common in otherwise healthy chil-
rhages in infants are due to abuse (Matschke et al., 2009). In dren with arterial ischemic stroke (Ganesan et al., 2003). At
older children and adults, low admission Glasgow Coma Scale least 10% of hemorrhagic strokes and most subarachnoid
(GCS) score, low systolic blood pressure, brain herniation, hemorrhages in children are caused by ruptured aneurysms
and requirement for decompressive craniotomy are all predic- (Jordan et al., 2009).
tors of post-traumatic cerebral infarction (Tian et al., 2008). One potential cause for stroke in pediatric patients is
Bony abnormalities of the vertebrae or abnormalities of vessel moyamoya disease. Moyamoya is a rare, chronic, progressive
walls due to collagen-vascular disease or metabolic disease steno-occlusive intracranial vasculopathy involving the distal
may predispose to cervicocephalic arterial dissection after supraclinoid internal carotid artery and the proximal anterior
mild trauma. Arterial dissection may be idiopathic in and middle cerebral arteries, and is associated with the forma-
otherwise apparently normal children (Rafay et al., 2006). A tion of an abnormal vascular network at the base of the brain
prothrombotic state associated with trauma can promote resembling a “puff of smoke” on angiography. The mechanism
thrombosis and worsen outcome; trauma patients who go of the disease is still unknown. Moyamoya disease has a high
into disseminated intravascular coagulation have worse out- prevalence in Japan, Korea, and China but has been reported
comes than those who do not. Trauma is the most common worldwide (Han et al., 2012). There is a bimodal distribution,
precipitant of hemorrhages in children with hemophilia. In presenting most often in children (girls more common
neonates, compression of the sagittal sinus due to head posi- than boys) younger than 10–15 years and in adults in the
tion has been associated with cerebral sinovenous thrombosis; third to fifth decades of life. Children usually present with
changing head position or minimizing compression with a recurrent transient ischemic attacks or strokes, headaches,
specialized pillow may improve venous flow (Tan et al., 2013). seizures, or movement disorders. Adults may present with
intracranial hemorrhage, including subarachnoid hemor-
rhage, subependymal hemorrhage, or intraventricular hemor-
Infection rhage. Some patients with moyamoya develop brain aneurysms.
The consequences of bacterial meningitis are disseminated Many disorders have been associated with moyamoya disease
intravascular coagulopathy and vascular inflammation, and (Table 68.1).
subsequent arterial or venous thrombosis and infarction. Fibromuscular dysplasia (FMD), a segmental, non-
Other cerebrovascular complications of meningitis include atherosclerotic, noninflammatory vascular disease of unknown
vasculitis, vasospasm, intracranial aneurysm formation, and pathophysiology, may result in arterial stenosis, vessel occlu-
rarely subarachnoid hemorrhage. Group B streptococcal men- sion, aneurysm, and/or arterial dissection. FMD most com-
ingitis is an important cause of stroke in neonates and trans- monly affects adult women and is rarely recognized in children
mitted vertically from the mother or horizontally by nursery with cerebral ischemia (DiFazio et al., 2000; Kirton et al., 2013
staff. During the first 2 months of life, infants are susceptible Zurin et al., 1997).
to bacteria found in maternal flora or in the local environ- Primary angiitis of the central nervous system (PACNS)
ment, including group B Streptococcus, Gram-negative enteric may occur in children and can be fatal if not treated with
bacilli, and Listeria monocytogenes. After 2 months of age, Strep- aggressive immune suppression. There are more frequently
tococcus pneumoniae and Neisseria meningitides are the most reported cases of less virulent vasculopathies in children, often
common causes of bacterial meningitis. The institution of occurring after varicella infection, which respond to aspirin
Haemophilus influenzae type b vaccination at 2 months of age alone and do not require immune suppression (Chabrier
has led to a dramatic drop in H. influenzae-b meningitis. In et al., 1998; Lanthier et al., 2001). Stroke can result from vas-
immunosuppressed patients such as those with cancer or culopathy outside the brain. Childhood stroke can be a first
acquired immune deficiency syndrome (AIDS), Aspergillus manifestation of Takayasu arteritis, an inflammatory large-
species may lead to vasculitis and infarction. There are numer- vessel vasculitis that affects the aorta and its branches (Brunner
ous possible causes of ischemic and hemorrhagic stroke in et al., 2008). A growing body of literature exists examining the
HIV/AIDS. Patients with AIDS may develop marantic endocar- role of inflammatory factors in stroke in children and in
ditis, arteriopathy of medium and small vessels or aneurysms, adults.
and although the presumed cause for most cases is direct or Although true migrainous infarctions are rare, migraine is
secondary infection, the exact pathophysiology is not always associated with a twofold increased risk of ischemic stroke.
clear. Highly active antiretroviral therapy for AIDS may result The risk is more apparent for individuals who have migraine
in dyslipidemia and may itself cause vascular injury and accel- with aura, smokers, and women who use oral contraceptives
erated atherosclerosis (Mondal et al., 2004). Tuberculosis (Shürks et al., 2009). While migraine has been associated
leads to meningitis in 1% to 2% of cases, which may cause with childhood stroke in large studies (Gioia et al., 2012;
vasculitis and infarction (Starke, 1999). Several cases of stroke Pezzini et al., 2014), a prospective study of 1008 children with
occurred in children with neurobrucellosis (Salih et al., migraine did not detect any clear infarcts (Mar et al., 2013).
2006b). Lyme disease is a rare cause of infectious vasculitis
and aneurysm formation. Varicella-zoster virus (VZV) may
cause vasculitis by direct infection of the arterial wall or by a
Drugs/Toxins
postinfectious inflammatory reaction that manifests weeks to Abuse of illicit drugs is an important cause of ischemic and
months after the primary infection. Even minor recent infec- hemorrhagic strokes in young patients. Maternal use of cocaine
tion has been identified as a risk factor for stroke, and may act may lead to vasospasm and cerebral infarction in the fetus,
by inducing an inflammatory prothrombotic state or by and use of cocaine by children may lead to intracranial hemor-
causing vascular injury (Hills et al., 2012). In some cases, rhage or ischemic stroke. Other drugs such as amphetamines,
arteriopathy is associated with recent upper respiratory infec- which lead to sudden increases in blood pressure or vaso­
tion (Amlie-Lefond et al., 2009a). spasm, also raise the risk of infarction. Ischemic stroke has
Stroke in Children 1001

TABLE 68.1  Disorders Associated with Moyamoya Other metabolic diseases lead to cerebral infarction by con-
tributing to thrombosis from arterial damage. Homocystinuria 68
Neonatal anoxia Brain tumors
may lead to infarction, presumably through elevated homo-
Head trauma Parasellar tumors cysteine levels and subsequent vascular injury. The C677T
Basilar meningitis Wilms tumor MTHFR gene variant may be associated with childhood stroke,
but it is not clear how large a role it plays; at least 10% of
Leptospirosis Post-radiation vasculopathy healthy children are homozygous for the MTHFR C677T
Tuberculosis Atherosclerotic disease (Gunther et al., 2000; Koch et al., 1999; Nowak-Göttl et al.,
Neurofibromatosis 1 Hypertension
1999). Studies vary on the degree of associated risk, and car-
riers do not always have elevated homocysteine levels at the
Tuberous sclerosis complex Cerebral dissecting and time of infarction. Fabry disease is an X-linked lysosomal
saccular aneurysms storage disease that causes a deficiency of α-galactosidase and
Sturge–Weber syndrome Fibromuscular dysplasia resultant accumulation of glycolipids in the endothelial wall.
Phakomatosis pigmentovascularis Coarctation of the aorta
Male patients experience paresthesias of the hands and feet
type IIIB and cardiac abnormalities that can begin in childhood; hypo-
hidrosis and renal dysfunction tend to occur later in the course
Pseudoxanthoma elasticum Renal artery stenosis of the disease (Ries et al., 2005). Both male and female het-
Hypomelanosis of Ito Arteriovenous malformations erozygotes are susceptible to cerebral thrombosis, possibly
Marfan syndrome Cavernous malformations
because of an increase in vasoreactivity in damaged vessels or
endothelial and leukocyte activation. Males may be more
Turner syndrome Systemic lupus erythematosus severely affected but rarely show cerebrovascular involvement
Williams syndrome Sneddon syndrome before age 23 (Schiffmann, 2001). α1-Antitrypsin deficiency
may lead to decreased structural integrity of the arterial wall
Noonan syndrome Polyarteritis nodosa
by disrupting the balance of activity between proteases and
Prader–Willi syndrome Sjögren symdrome antiproteases. α1-Antitrypsin deficiency has been associated
Alagille syndrome Sarcoidosis with aneurysms and with vascular changes consistent with
fibromuscular dysplasia. The hyperlipidemias can cause
Apert syndrome Use of oral contraceptives atherosclerotic vascular changes in children similar to those
Down syndrome Drug abuse (cocaine) in older adults.
Sickle cell anemia Homocystinuria
β-Thalassemia Type I glycogenosis Gender and Ethnicity
Fanconi anemia Osteogenesis imperfecta The role of gender in pediatric stroke has been controversial.
Aplastic anemia Glycogen storage disease One study found that both arterial ischemic stroke and sino-
Hereditary spherocytosis Hyperlipoproteinemia
venous thrombosis are more common in boys, with male
predominance particularly in cases of stroke caused by trauma-
Protein C deficiency Primary oxalosis associated arterial dissection (Golomb et al., 2009). However,
Protein S deficiency Hirschprung disease a recent population-based study in England did not find male
gender to be a risk factor (Mallick et al., 2014). Higher testo-
Antiphospholipid antibody Giant cervico-facial
syndrome (APAS) hemangiomas
sterone levels have been associated with increased risk of
stroke in boys (Normann et al., 2009).
Thrombotic thrombocytopenic Hyperthyroidism Ethnicity is a risk factor for stroke. Black children appear
purpura to be at an increased risk for both ischemic and hemorrhagic
Factor XII deficiency Graves disease stroke which cannot be fully explained by sickle cell disease.
Asian ethnicity has been associated with stroke risk in the
Adapted from K. Houkin and T. Mikami, Moyamoya Disease and
Moyamoya Syndrome, in: J.E. Wanebo, N. Khan, J.M. Zabramski,
United Kingdom but not in the United States; differences in
R.F. Spetzler (Eds), Moyamoya Disease: Diagnosis and Treatment. the ethnic makeup of the Asian populations between coun-
Thieme, New York, 2011. tries may explain some of the difference (Fullerton et al.,
2003; Mallick et al., 2014).
Gender and ethnicity may influence recovery from stroke.
A study of mortality from childhood stroke in England and
Wales from 1921 to 2000 found that boys had a higher mortal-
ity rate (Mallick et al., 2009). Black children have had higher
been associated with both “natural” and “synthetic” (“Spice”) stroke mortality than white children in the United States. This
marijuana use. Proposed mechanisms include cannabinoid- has been in part due to sickle cell anemia, and the disparity
induced intracranial arteriopathy and/or cardiovascular effects has decreased since the Stroke Prevention Trial in Sickle Cell
such as arrhythmias (Barber et al., 2013; Freeman et al., 2013; Anemia (STOP trial) (Lehman et al., 2013).
Wolff et al., 2011). Accidental ingestion or overdose of medica-
tions used to treat thrombosis may lead to hemorrhage.
Genetic
Metabolic Genome-wide association studies and whole-exome sequenc-
The mitochondrial diseases may lead to metabolic strokes, ing have led to the identification of genetic risk factors, but
particularly during times of metabolic stress. MRI can demon- the function of the identified genes is not always clear. Muta-
strate infarction in nonvascular territories (Fig. 68.5). However, tions in the CECR1 (cat eye syndrome chromosome region,
in rare cases, mitochondrial mutations have been associated candidate 1) which encode adenosine deaminase 2 have
with moyamoya vasculopathy (Longo et al., 2008; Papavasil- been associated with early stroke and vasculopathy. Zebrafish
iou et al., 2007). and human studies suggest that adenosine deaminase 2 may
1002 PART III  Neurological Diseases and Their Treatment

A B C
Fig. 68.5  Magnetic resonance imaging (MRI) of an 18-year-old girl who presented at age 12 with headaches and went on to develop
bilateral incoordination, decreased attention span, decline in school performance, and fatigability. A, Fluid spin echo inversion recovery
MRI demonstrates multiple bilateral lesions in nonvascular territories. B, A section of the involved area is selected for evaluation using MR spec-
troscopy (MRS). C, MRS demonstrates a lactate doublet peak consistent with mitochondrial disease in an area of signal abnormality (arrows).

function as a growth factor for endothelial cell development encephalopathy (Salpietro V. et al., 2014) are all causes of
(Zhou et al., 2014). The AB collagen type IV alpha (COL4A1) sudden onset of focal or multifocal neurological symptoms,
gene codes for a collagen which plays a role in the basement and all may cause multiple T2 bright lesions on MRI.
membranes of cerebral vasculature, and mutations can lead to
hemorrhagic strokes starting in childhood (Jeanne et al., 2012;
Shah et al., 2010). Genetic variants in the plasma glutathione
EVALUATION
peroxidase gene (GPX3), which codes an enzyme which may History and Physical
modify fibrinogen, has been associated with pediatric ischemic
In the young patient, the history should include questions
stroke due to arteriopathy (Nowak-Göttl et al., 2011). A pre-
about delivery and the perinatal period, attainment of hand
disposition to pediatric stroke has been associated with four
preference, and basic developmental milestones. Develop-
members of the ADAMTS gene family: ADAMTS13, ADAMTS17,
ment of a hand preference before 1 year of age may be a sign
ADAMTS2, and ADAMTS12. The currently best-understood
of a mild hemiparesis in the nondominant hand, which could
of these genes, ADAMTS13, codes for von Willebrand factor
be due to a perinatal infarction. Any early hemorrhagic or
cleaving protease, and leads to increased von Willebrand
ischemic infarction may lead to slowed development. Medical
factor-induced endothelial platelet aggregation. Some muta-
history should include questions about previous hemor-
tions in this gene are associated with familial thrombocyto-
rhages, abnormal bruising, petechiae, thromboses, and other
penic purpura. However, the biological functions of the
medical conditions that may raise the risk of early stroke (e.g.,
ADAMTS genes with the greatest risk for stroke, ADAMTS2 and
complex congenital heart disease, renal failure, sickle cell
ADAMTS12, are unknown (Arning et al., 2012).
anemia). Children with moyamoya usually present with recur-
rent transient ischemic attacks or cerebral infarction. Symp-
toms may be triggered by crying, physical exertion, or
DIFFERENTIAL DIAGNOSIS hyperventilation. Family history should include questions
Children with stroke often present with seizures, and in the first about abnormal bleeding in other family members, strokes or
few hours after a seizure, before cranial imaging, it can be dif- heart attacks before age 45, peripheral arteriopathy, or deep
ficult to determine whether a new hemiparesis is due to a venous thrombosis. A history of multiple miscarriages may be
temporary postictal Todd paresis or to infarction. Todd paresis suggestive of antiphospholipid antibody syndrome.
usually does not last more than 24 hours, though in rare cases Physical examination should include examination of the
it may persist several days. Migraine may lead to infarction and face for signs of dysmorphic features suggestive of a genetic
permanent motor impairment, but hemiplegic migraine may syndrome. Always assess head circumference. Early stroke may
lead to temporary motor impairment. A strong family history lead to macrocephaly due to hydrocephalus or to microceph-
of hemiplegic migraine or documentation of a missense muta- aly due to tissue loss and poor brain growth from infarction.
tion of the CACNA1A calcium channel gene on chromosome Examination of the skin should document signs of bruising
19p13 may help differentiation (Terwindt et al., 2002), but or petechiae, livedo reticularis suggestive of Sneddon syn-
variation exists in mutations among families (see Chapter 103). drome, systemic lupus erythematosus or other autoimmune
Alternating hemiplegia of childhood is a progressive neurode- disorders, café-au-lait spots suggestive of neurofibromatosis
generative condition. The origin is generally unclear, although type 1, hypopigmented macules suggestive of tuberous sclero-
cerebrovascular factors and mitochondrial disease may be con- sis complex, excess skin laxity suggestive of a collagen disorder
tributory. Neuroimaging studies usually do not demonstrate such as Ehlers–Danlos syndrome, cyanosis suggestive of heart
pathology. Edema, bleeding, or shifting of brain tumor may failure and anoxia, and pallor suggestive of anemia. The head
cause sudden onset of neurological signs. Encephalitis or and neck should be auscultated for vascular bruits and the
meningoencephalitis may lead to sudden onset of focal neuro- heart for murmurs and arrhythmias; peripheral pulses should
logical symptoms. Several metabolic diseases including glutaric be compared, and the abdomen should be auscultated for
aciduria and carbohydrate-deficient glycoprotein syndrome (see renal bruits suggestive of a systemic vascular disorder. Neuro-
Chapter 91) can present with stroke-like episodes, and serum logical examination should look for signs of focal abnormal-
and urine testing together with MRI help make the diagnosis. ity, with the caveat that signs may not localize well in the very
Acute disseminated encephalomyelitis (ADEM), multiple scle- young child or may be difficult to assess in the frightened or
rosis, vasculitis, and the vasculitic form of Hashimoto uncooperative child (Table 68.2).
Stroke in Children 1003

TABLE 68.2  Physical Examination of the Child with Stroke


68
Finding Possible significance/suggestive of Finding Possible significance/suggestive of
HEAD CIRCUMFERENCE Telangiectasias Osler–Weber–Rendu disease (hereditary
hemorrhagic telangiectasia)
Macrocephaly Hydrocephalus caused by IVH, SDH,
SAH, or vascular malformation Oral/genital ulcers Behçet disease
Microcephaly Failure of brain growth resulting from Angiokeratomas Fabry disease
stroke or genetic disorder
Lentigines in non-sun- Predisposition to dissection or to atrial
EYES exposed areas myxoma, which may lead to
cardioembolic stroke
External/Iris
Discoloration of fingers with Raynaud phenomenon as sign of
Epicanthal folds, Brushfield Down syndrome
cold: white, blue, then collagen disease or systemic lupus
spots
red erythematosus
Horner syndrome Carotid dissection (postganglionic)/
Subcutaneous nodules on Rheumatic fever, systemic lupus
vertebral dissection (central)
elbows, forehead, erythematosus, rheumatoid arthritis
Pulsating exophthalmos Carotid-cavernous fistula tendons
Lens subluxation Marfan syndrome, homocystinuria Erythematous macules that Acute varicella (chicken pox); shingles if
evolve to clear, fluid-filled in a dermatomal distribution
Angioid streaks Pseudoxanthoma elasticum
vesicles
Xanthelasma on lids, Hyperlipidemia
Multiple round, white, Past varicella infection
corneal arcus
puckered scars
Corneal opacity Fabry disease
Needle tracks IV drug addiction with risk for
Retina endocarditis and HIV
Papilledema Increased ICP resulting from MOUTH
hydrocephalus, vascular malformation,
High, arched palate Marfan syndrome
or acute brain edema
Petechial hemorrhages Infective endocarditis
Hemorrhages Trauma (consider child abuse), bleeding
diathesis, ruptured aneurysm, collagen HEART AND PERIPHERAL PULSES
disease, emboli
Murmur Complex congenital heart disease, valve
Vasculopathy Systemic vasculitis abnormality
Angioid streaks Pseudoxanthoma elasticum, Paget Decreased pulses Takayasu arteritis
disease, sickle cell anemia
Increased pulses Hypertension
Angioma Familial cavernous angiomatosis, von
ABDOMEN
Hippel–Lindau disease
Hepatomegaly Infection, cancer, liver failure
SKIN
Bruit Renal artery stenosis
Bruising Bleeding diathesis (consider child abuse)
BACK
Petechiae Platelet count low or dysfunction; DIC
Scoliosis, vertebral Possible increased risk of dissection
Purpura Henoch–Schönlein purpura
anomalies
Pallor Anemia
HANDS
Erythema Polycythemia
Long, tapering fingers Marfan syndrome
Cyanosis Complex congenital heart disease, other
Other anomalies of bones May be seen in association with
cause of hypoxia
of hands congenital heart disease
Skin necrosis Meningococcemia
Clubbing of fingers Congenital heart disease
Café-au-lait spots Neurofibromatosis type 1
JOINTS
Hypopigmented macules, Tuberous sclerosis complex
Painful, restricted Arthritis due to autoimmune disease,
shagreen patches, facial
movement past bleeds (hemophilia)
angiofibromas
Warm Autoimmune disease, infection
Yellow papules Pseudoxanthoma elasticum
OVERALL SIZE
Premature aging Progeria
Tall Marfan syndrome, homocystinuria
Malar rash Systemic lupus erythematosus
Short Progeria, mitochondrial disease,
Skin laxity Ehlers–Danlos syndrome type IV
dwarfism (risk of vertebral anomalies)
DIC, Disseminated intravascular coagulation; HIV, human immunodeficiency virus; ICP, increased intracranial pressure; IV, intravenous; IVH,
intraventricular hemorrhage; SAH, subarachnoid hemorrhage; SDH, subdural hematoma.
Physical findings taken from Hurst, J.W., 1993. Cardiovascular Diagnosis: The Initial Examination. Mosby-Year Book, St. Louis. Information on
lentigines from Neau, J.P., Rosolacci, T., Pin, J.C., et al., 1993. Cerebral infarction, cardiac myxoma and lentiginosis. Rev Neurol (Paris) 149,
289–291; and Schievink, W.I., Michels, V.V., Mokri, B., et al., 1995. Brief report: a familial syndrome of arterial dissections with lentiginosis. N Engl
J Med. 332, 576–579.
1004 PART III  Neurological Diseases and Their Treatment

Imaging Studies remains the gold standard, and should include selective injec-
tions of both ICAs, external carotid arteries, and vertebral
Ultrasound (see Chapter 40) is useful in assessing IVH or arteries. Use all forms of neuroimaging in combination with
periventricular leukomalacia in the premature infant or carotid other laboratory and physical findings, because no one tech-
flow in any child, but its sensitivity for detecting arterial stroke nique has perfect sensitivity or specificity for detecting vascu-
in the neonate is probably below 50% (Golomb et al., 2001). lopathy. Continuous arterial spin labeling MRI has been used
Power Doppler ultrasound may be useful in detecting CVT in in research settings to study regional cerebral blood flow in
the neonate. Transcranial Doppler ultrasound is used to screen persons with sickle cell anemia (Van den Tweel et al., 2009).
children with sickle cell anemia, and higher blood flow veloci- Other newer forms of imaging such as functional MRI and
ties are associated with higher stroke risk. Studies have sug- diffusion-weighted tensor imaging are used in research set-
gested that this screening should begin in infancy (Telfer et al., tings and may provide insights into stroke evolution and
2007). recovery but are not widely available.
Cranial computed tomography (CT) (see Chapter 39) is
better at assessing hemorrhage in the older child and ischemic
stroke in the neonate and older child, but it may not detect Coagulation Workup
arterial stroke until 24 hours or more after the event. CT angi- The basic evaluation for prothrombotic disorders may include
ography and venography (see Chapter 40) can image the cer- prothrombin time (PT) with international normalized ratio
ebral vasculature. (INR) and activated partial thromboplastin time (aPTT) and
MRI is the imaging tool of choice for most types of child- a complete blood cell count, including platelets, protein C,
hood stroke, and diffusion-weighted imaging (DWI) can protein S and antithrombin levels, activated protein C resist-
detect brain ischemia within hours (see Chapter 39). DWI ance, plasminogen, fibrinogen, homocysteine, antiphospholi-
stays bright for approximately 2 weeks in the older child pid antibody screen, lipoprotein (a), and a lipid panel. Testing
or adult but may “normalize” (pseudonormalization) within for protein Z may become part of the evaluation in the future
days in the neonate (Mader et al., 2002). Stroke imaging at if other studies confirm its role in childhood stroke. Genetic
any age should include DWI, T2, and fluid-attenuated inver- testing may include screening for the factor V Leiden muta-
sion recovery (FLAIR) to detect areas of early infarction. tion, the prothrombin 20210A gene, and the MTHFR gene
Magnetic resonance angiography (MRA) may be helpful in variants. Some mutations are very common, and the ratio of
diagnosing vasculitis and dissection involving larger vessels. symptomatic to nonsymptomatic carriers may be very high in
In moyamoya, MRI typically demonstrates diminished flow some populations; more than 40% of healthy children in a
voids in the distal ICAs, and proximal MCAs and ACAs, in European study were either homozygous or heterozygous for
addition to prominent collateral flow voids in the basal the C677T MTHFR variant (Koch et al., 1999). The frequency
ganglia region (Fig. 68.6). Magnetic resonance spectroscopy of different prothrombotic risk factors varies among popula-
(MRS) and single-photon emission CT (SPECT) demonstrate tions (Salih et al., 2006a). Unfortunately, identifying asymp-
changes in regional blood flow and may be helpful in assess- tomatic carriers of prothrombotic genes might adversely affect
ing patients with moyamoya disease and other sources of later health, life, and disability insurance status (Golomb
vasculopathy. MRS provides the earliest detection of ischemic et al., 2005).
lesions. Conventional angiography can clarify the structure of
vascular malformations and is the most accurate method for
detecting moyamoya, vasculitis, and dissection, but angiogra-
Cardiac Evaluation
phy may not always be possible in the acutely ill child. In The basic cardiac evaluation may include electrocardiogram
patients suspected of moyamoya, six-vessel angiography (ECG) with rhythm strip and a transthoracic echocardiogram

A B

C D

E F
Fig. 68.6  A 14-month-old girl with a history of complex partial seizures now develops a postictal right hemiparesis. A–F, DWMRI shows
acute transcortical infarcts involving the right MCA territory, small segment of the left ACA territory, small segmental left PCA territory, and punctate
left MCA territory. On MRA, the intracranial arterial circulation demonstrates left supraclinoid carotid occlusion and reconstitution of left M1 and
A1 segments via moyamoya-type collaterals. There was normal course and caliber of both cervical carotid and vertebral arteries.
Stroke in Children 1005

with injection of agitated saline to screen for a patent foramen anticoagulated are at increased risk for thrombus propagation,
ovale. If any suspicion exists for cardioembolic events and which can be asymptomatic and lead to poor outcome. Other 68
those two studies are unrevealing, perform transesophageal low-molecular-weight heparinoids such as danaparoid may be
echocardiography and a Holter monitor study. alternatives for patients with heparin-induced thrombocyto-
penia (Neuhaus et al., 2000; Ranze et al., 1999; Risch et al.,
2006).
Other Studies In sickle cell anemia, exchange transfusion is the usual
Flexion and extension radiographs of the cervical spine may treatment for acute stroke (Menkes et al., 2000). The Stroke
identify bony abnormalities predisposing to dissection. Elec- Prevention in Sickle Cell Trial (STOP) demonstrated that in
troencephalogram (EEG) may help localize the lesion in chil- children with vasculopathy shown by transcranial Doppler
dren who present with seizures and is helpful for evaluating ultrasound screening, regular transfusions to keep the sickle
cerebral function in the unresponsive and possibly locked-in hemoglobin concentration at less than 30% reduced the risk
patient with a brainstem stroke (see Chapter 5). Visual evoked of recurrent stroke by 90% (Adams, 2000). This trial has
potentials and brainstem auditory evoked potentials may be helped decrease the disparity in stroke mortality between
particularly helpful in evaluating the very young or somnolent black children and white children in the United States
patient (see Chapter 34). (Lehman et al., 2013). However, chronic transfusions carry the
Serum and plasma studies may help identify the cause of risk of infection and cause increases in serum ferritin (Files
the stroke. Thrombocytopenia, deficiencies of factors I, VII, et al., 2002). Some children require treatment with deferox-
VIII, IX, and XIII, and deficiency of von Willebrand factor are amine chelation to prevent iron overload (Wayne et al., 2000).
all risk factors for intracranial hemorrhage, whereas poly- Stopping exchange transfusion therapy leads to increased vas-
cythemia, thrombocythemia, and anemia are all risk factors cular disease and stroke risk (Adams and Brambilla, 2005).
for ischemic stroke. Abnormalities in platelet count and fibrin- Attempts at treating and preventing metabolic stroke and
ogen may be markers for disseminated intravascular coagula- cerebrovascular disease have been made by addressing the
tion, which may lead to thrombosis and ischemic stroke. pathological defects in metabolic pathways, using coenzyme
Elevated serum lactate is a marker for mitochondrial disease, Q10 and other antioxidant vitamins. L-Arginine improves
but not all patients with mitochondrial disease have elevated endothelial dysfunction in children with MELAS (mitochon-
serum lactate; genetic tests may be required to confirm the drial encephalopathy, lactic acidosis, and strokes) and may aid
diagnosis. Muscle biopsy may help confirm mitochondrial in treating the stroke-like episodes (Koga et al., 2006). Folate,
disease (see Chapter 93). Serum studies for plasma vitamin B6, and vitamin B12 all play a role in homocysteine
α-galactosidase will identify Fabry disease. Serum ammonia, metabolism, which is impaired in homocystinuria and in
amino acids, and organic acids may help identify metabolic carriers of the MTHFR C677T gene variant. Several different
disease such as hyperhomocysteinemia and mitochondrial enzyme abnormalities may lead to homocystinuria, and the
diseases that can lead to stroke (Menkes, 2000). enzyme affected determines whether supplementation with
folate, vitamin B6, or vitamin B12 is helpful (Menkes, 2000).
Dietary supplementation with folate lowers serum homo-
TREATMENT cysteine levels in adults with the MTHFR C677T gene variant
(Thuillier et al., 1998) and may be helpful in children with it.
The Acute Period and Initiating Chronic Therapy Enzyme replacement with α-galactosidase decreases the symp-
Children with intracranial hemorrhage or hemorrhagic stroke toms of Fabry disease and normalizes cerebrovascular reactiv-
require close observation in the first hours. Children with ity but does not remove the risk of recurrent stroke (Schiffmann
hemophilia need immediate factor replacement and may et al., 2006).
require blood transfusion. Children with large hematomas Patients with protein C deficiency have been treated with
with significant mass effect may need surgery (Johnson, 2000). protein C concentrate (Ettingshausen et al., 1999). Good
Any child presenting with unexplained or poorly explained results are reported when using antithrombin concentrate to
intracranial hemorrhage should also be evaluated for other treat consumptive coagulopathy and antithrombin deficiency
sites of injury and possible child abuse. at the time of acute stroke, but it does not appear to prevent
Between 2000 and 2003, fewer than 2% of children with thrombosis in leukemia patients treated with L-asparaginase
ischemic stroke received thrombolytic therapy (Janjua et al., (Hongo et al., 2002).
2007). Some of these children were not treated in accordance
with guidelines used for adults (Amlie-Lefond, 2009b; Roach
et al., 2008). Few children present within the originally man-
Additional Issues in Chronic Therapy
dated 3 hours of infarct onset. The benefit of thrombolytic Patients with hemophilia and severe bleeding may require
therapy in children is not clear. No evidence-based guidelines prophylaxis with regular factor transfusions. Young or ataxic
are currently available on using intravenous or intra-arterial children may need to wear protective helmets until their
thrombolytics in children. Unfortunately, a recent NIH-funded balance improves.
dose-finding study of tissue plasminogen activator for acute Patients with genetic chronic thrombotic abnormalities
pediatric stroke was closed because of difficulty enrolling such as protein C or S deficiency, or with antiphospholipid
patients (Amlie-Lefond, 2014). Thrombolytic therapy may not antibody syndrome, may require long-term care with low-
be helpful in neonates, who have lower levels of plasminogen molecular-weight heparin or warfarin (Andrew et al., 2000).
than older children (Andrew et al., 2000). For many patients, however, the best long-term therapy is not
No clear guidelines are available on the use of heparin in clear. Low-molecular-weight heparin may be used for 3 to
pediatric arterial stroke and CVT patients either. Pilot studies 6 months after cervicocephalic arterial dissection or CVT
and case series have described good results with heparin and (Andrew and deVeber, 1999), with an Insuflon patch to make
low-molecular-weight heparin (deVeber et al., 1998; Massi- administration tolerable. Warfarin or aspirin are more often
cotte et al., 1996). Large-scale studies with catheter-related used for long-term therapy (Andrew et al., 2000). Families of
noncranial thrombosis have shown good results (Andrew children on warfarin need to be counseled about regulating
et al., 2000). Moharir et al. (2009) showed that neonates vitamin K levels in the diet, bony changes, possible tera-
and children with sinovenous thrombosis who are not togenicity, and the risks of under- or overtreatment. Families
1006 PART III  Neurological Diseases and Their Treatment

of children taking aspirin are generally concerned about Reye families, preferably before adolescence, as some oral contra-
syndrome, but there are no reports in children taking aspirin ceptives are used to treat acne or menstrual disorders in young
for stroke prophylaxis (Roach, 2000). Strater and colleagues adolescent women.
(2001) did a prospective follow-up study of 135 children with
first onset of ischemic stroke. The children received prophy-
lactic treatment with either low-dose low-molecular-weight
SUMMARY
heparin or aspirin and were followed for a median of 36 Children with stroke are at risk for future cognitive impair-
months. Low-dose low-molecular-weight heparin was not ment, motor impairment, and epilepsy. In full-term neonates,
superior to aspirin in preventing stroke recurrence. For chil- it is difficult to predict outcome based on initial cranial
dren with sickle cell anemia, chronic treatment with hydrox- imaging, and it is unclear whether the side on which the inf-
yurea can increase levels of fetal hemoglobin and decrease arcts occur affects language development. Evidence of degen-
complications of sickle cell anemia, but careful monitoring is eration of corticospinal tracts and resulting asymmetry in the
required for cytotoxicity and genotoxicity (Khayat et al., midbrain on MRI performed during the second 6 months of
2006). Bone marrow transplantation with matched donors life does predict hemiplegia. About half of children with neo-
may be an option for some children (Vermylen, 2003). Surgery natal arterial ischemic stroke have feeding problems; when
or interventional procedures play a role in treating some types severe, feeding dysfunction can lead to aspiration and failure
of cerebrovascular disease. Indirect surgical revascularization to thrive (Barkat-Masih et al., 2010). Children who present
procedures, such as encephaloduroarteriosynangiosis (EDAS), with signs of perinatal stroke after the perinatal period appear
encephalomyosynangiosis (EMS), encephalomyoarteriosy- to have worse motor outcomes than children with perinatal
nangiosis (EMAS), pial synangiosis, multiple burr holes, crani- stroke who present in the neonatal period (Lee et al., 2005).
otomy with inversion of the dura, omental transplantation, For these children, young age at presentation, fever at presen-
omental pedicle graft, and cervical sympathectomy, may be tation, and right middle cerebral artery infarction have been
used to treat children with moyamoya disease. Direct revascu- associated with poor outcome (Cnossen et al., 2010). Bilateral
larization procedures such as superficial temporal artery (STA) hemispheric lesions and low muscle tone of affected limbs at
to middle cerebral artery (MCA) bypass are technically diffi- presentation have also been associated with poor outcome
cult to perform in children. Other surgical procedures such as (Kim et al., 2009). Studies in both infants and older children
clipping, coiling, radioablation, or removal may be useful in show that larger and multiple infarcts are more likely to cause
the treatment of aneurysms or malformations. cognitive and motor impairment as well as epilepsy. Overall,
Socioeconomic status and social stressors may play a role the intelligence of children with stroke falls within the normal
in family compliance with therapy and patient outcome. In a range, but some require extra help in school.
Texas study of children with sickle cell anemia, families with The prognosis for children with progressive cerebrovascular
private insurance were more likely to be compliant with diseases is particularly problematic. Those children with severe
Doppler ultrasound screenings than families on Medicaid forms of sickle cell or moyamoya disease who are not treated
(Raphael et al., 2008). or who do not respond to therapy may develop significant
cognitive and motor disabilities and epilepsy (Lagunju et al.,
2013; Weinberg et al., 2011). More than 20% of children who
Other Issues survive childhood arterial ischemic stroke may have recurrent
The risks that accompany pregnancy in adolescence are greater transitory ischemic attacks or stroke (Ganesan et al., 2002).
for young women with a history of hemorrhagic or ischemic Prothrombotic disorders and abnormalities on vascular
stroke (see Chapter 112). imaging raise the risk of recurrence (Strater et al., 2002).
Oral contraceptives carry a very small risk for the general
population but may carry more risk for women with pro-
thrombotic disorders (Slooter et al., 2005). Oral contraceptive REFERENCES
use raises the risk of both arterial thromboembolism and CVT. The complete reference list is available online at https://expertconsult
These issues require explicit discussion with patients and their .inkling.com/.
Stroke in Children 1006.e1

sible MTHFR and eNOS gene-gene interplay? J. Child Neurol. 24


REFERENCES (7), 823–827. 68
Adams, R.J., 2000. Lessons from the Stroke Prevention Trial in Sickle Domi, T., Edgell, D.S., McCrindle, B.W., et al., 2008. Frequency, pre-
Cell Anemia (STOP) study. J. Child Neurol. 15, 344–349. dictors, and neurologic outcomes of vaso-occlusive strokes associ-
Adams, R.J., Brambilla, D., 2005. Discontinuing prophylactic transfu- ated with cardiac surgery in children. Pediatrics 122 (6),
sions used to prevent stroke in sickle cell disease. N. Engl. J. Med. 1292–1298.
353, 2769–2778. Ettingshausen, C.E., Veldmann, A., Beeg, T., et al., 1999. Replacement
Amlie-Lefond, C. 2014. Thrombolysis in Pediatric Stroke Study (TIPS) therapy with protein C concentrate in infants and adolescents with
5R01NS065818-03. http://projectreporter.nih.gov/project_info_ meningococcal sepsis and purpura fulminans. Semin. Thromb.
News.cfm?aid=8544509&icde=19896749 Status update was per- Hemost. 25, 537–541.
sonal and group communication from PI at International Stroke Files, B., Brambilla, D., Kutlar, A., et al., 2002. Longitudinal changes
Study Meeting at the International Stroke Conference, February in ferritin during chronic transfusion: a report from the Stroke
2014. Prevention Trial in Sickle Cell Anemia (STOP). J. Pediatr. Hematol.
Amlie-Lefond, C., Bernard, T.J., Sebire, G., et al., 2009a. Predictors of Oncol. 24, 284–290.
cerebral arteriopathy in children with arterial ischemic stroke: Fitzgerald, K.C., Williams, L.S., Garg, B.P., et al., 2006. Cerebral
results of the International Pediatric Stroke Study. Circulation 119 sinovenous thrombosis in the neonate. Arch. Neurol. 63,
(10), 1417–1423. 405–409.
Amlie-Lefond, C., deVeber, G., Chan, A.K., et al., 2009b. Use of Fluss, J., Geary, D., deVeber, G., 2006. Cerebral sinovenous thrombosis
alteplase in childhood arterial ischaemic stroke: a multicentre, and idiopathic nephrotic syndrome in childhood: report of four
observational, cohort study. Lancet Neurol. 8 (6), 530–536. new cases and review of the literature. Eur. J. Pediatr. 165,
Andrew, M., deVeber, G., 1999. Pediatric Thromboembolism and 709–716.
Stroke Protocols. B. C. Decker, Hamilton, Ontario, Canada. Freeman, M.J., Rose, D.Z., Myers, M.A., et al., 2013. Ischemic stroke
Andrew, M., Monagle, P., Brooker, L., 2000. Thromboembolic Com- after use of the synthetic marijuana “spice”. Neurology 81 (24),
plications During Infancy and Childhood. B.C. Decker, London. 2090–2093.
Arning, A., Hiersche, M., Witten, A., et al., 2012. A genome-wide asso- Fullerton, H.J., Adams, R.J., Zhao, S., et al., 2004. Declining stroke
ciation study identifies a gene network of ADAMTS genes in the rates in Californian children with sickle cell disease. Blood 104,
predisposition to pediatric stroke. Blood 120 (26), 5231–5236. 336–339.
Barber, P.A., Pridmore, H.M., Krishnamurthy, V., et al., 2013. Canna- Fullerton, H.J., Wu, Y.W., Zhao, S., Johnston, S.C., 2003. Risk of stroke
bis, ischemic stroke, and transient ischemic attack: a case-control in children: ethnic and gender disparities. Neurology 61 (2),
study. Stroke 44 (8), 2327–2329. 189–194.
Barkat-Masih, M., Saha, C., Hamby, D.K., et al., 2010. Feeding prob- Gabis, L.V., Yangala, R., Lenn, N.J., 2002. Time lag to diagnosis of
lems in children with neonatal arterial ischemic stroke. J. Child stroke in children. Pediatrics 110, 924–928.
Neurol. 25 (7), 867–872. Gandhi, S.K., McKinney, J.S., Sedjro, J.E., et al., 2012. Temporal trends
Barrett, C.S., Bratton, S.L., Salvin, J.W., et al., 2009. Neurological in incidence and long-term case fatality of stroke among children
injury after extracorporeal membrane oxygenation use to aid pedi- from 1994 to 2007. Neurology 78 (24), 1923–1929.
atric cardiopulmonary resuscitation. Pediatr. Crit. Care Med. 10 (4), Ganesan, V., Chong, W.K., Cox, T.C., et al., 2002. Posterior circulation
445–451. stroke in childhood: risk factors and recurrence. Neurology 59,
Brunner, J., Armstrong, D., Feldman, B.M., et al., 2008. Childhood 1552–1556.
stroke as the presentation of Takayasu’s arteritis: diagnostic delay Ganesan, V., Prengler, M., McShane, M.A., et al., 2003. Investigation
can cause catastrophic complications. J. Rheumatol. 35 (6), of risk factors in children with arterial ischemic stroke. Ann. Neurol.
1228–1230. 53, 167–173.
Byrnes, J.W., Prodhan, P., Williams, B.A., et al., 2013. Incremental Gioia, L.C., Tollard, É., Dubuc, V., et al., 2012. Silent ischemic lesions
reduction in the incidence of stroke in children supported with the in young adults with first stroke are associated with recurrent stroke.
Berlin EXCOR ventricular assist device. Ann. Thorac. Surg. 96 (5), Neurology 79 (12), 1208–1214.
1727–1733. Giroud, M., Lemesle, M., Gouyon, J.B., et al., 1995. Cerebrovascular
Chabrier, S., Rodesch, G., Lasjaunias, P., et al., 1998. Transient cerebral disease in children under 16 years of age in the city of Dijon, France:
arteriopathy: a disorder recognized by serial angiograms in children a study of incidence and clinical features from 1985 to 1993. J. Clin.
with stroke. J. Child Neurol. 13, 27–32. Epidemiol. 48, 1343–1348.
Cnossen, M.H., Aarsen, F.K., Akker, S., et al., 2010. Paediatric arterial Golomb, M.R., Dick, P.T., MacGregor, D.L., et al., 2001. Cranial ultra-
ischaemic stroke: functional outcome and risk factors. Dev. Med. sound has a low sensitivity for detecting acute infarct in term
Child Neurol. 52 (4), 394–399. neonates (abstract). Ann. Neurol. 50, S120.
Cornelissen, E.A., Hirasing, R.A., Monnens, L.A., 1996. [Prevalence of Golomb, M.R., Fullerton, H.J., Nowak-Gottl, U., et al., 2009.
hemorrhages due to vitamin K deficiency in The Netherlands, Male predominance in childhood ischemic stroke: findings
1992–1994]. Ned. Tijdschr. Geneeskd. 140, 935–937. from the international pediatric stroke study. Stroke 40 (1),
DeBaun, M.R., Armstrong, F.D., McKinstry, R.C., Ware, R.E., Vichinsky, 52–57.
E., Kirkham, F.J., 2012. Silent cerebral infarcts: a review on a preva- Golomb, M.R., Garg, B.P., Walsh, L.E., et al., 2005. Perinatal stroke in
lent and progressive cause of neurologic injury in sickle cell anemia. baby, prothrombotic gene in mom: does this affect maternal health
Blood 119, 4587–4596. insurance? Neurology 65, 13–16.
Denier, C., Labauge, P., Bergamett, F., et al., 2006. Genotype-phenotype Gradnitzer, E., Urlesberger, B., Maurer, U., et al., 2002. [Cerebral hem-
correlations in cerebral cavernous malformations patients. Ann. orrhage in term newborn infants—an analysis of 10 years (1989-
Neurol. 60 (5), 550–556. 1999)]. Wien. Med. Wochenschr. 152, 9–13.
de Ribaupierre, S., Rilliet, B., Cotting, J., Regli, L., 2008. A 10-year Gunther, G., Junker, R., Strater, R., et al., 2000. Symptomatic ischemic
experience in paediatric spontaneous cerebral hemorrhage: which stroke in full-term neonates: role of acquired and genetic prothrom-
children with headache need more than a clinical examination? botic risk factors, [erratum appears in Stroke 2001 32, 279]. Stroke
Swiss Med. Wkly. 138 (5–6), 59–69. 31, 2437–2441.
deVeber, G., Andrew, M., Adams, C., et al., 2001. Cerebral sinovenous Han, C., Chen, J., Gao, S., Duan, L., 2012. Moyamoya disease in Asia.
thrombosis in children. N. Engl. J. Med. 345, 417–423. Neurol. Asia 17 (3), 175–181.
deVeber, G., Chan, A., Monagle, P., et al., 1998. Anticoagulation Hills, N.K., Johnston, S.C., Sidney, S., et al., 2012. Recent trauma and
therapy in pediatric patients with sinovenous thrombosis: a cohort acute infection as risk factors for childhood arterial ischemic stroke.
study. Arch. Neurol. 55, 1533–1537. Ann. Neurol. 72 (6), 850–858.
DiFazio, M., Hinds, S.R. 2nd, Depper, M., et al., 2000. Intracranial Hongo, T., Okada, S., Ohzeki, Z., et al., 2002. Low plasma levels of
fibromuscular dysplasia in a six-year-old child: a rare cause of child- hemostatic proteins during the induction phase in children with
hood stroke. J. Child Neurol. 15 (8), 559–562. acute lymphoblastic leukemia: a retrospective study by the JACLS.
Djordjevic, V., Stankovic, M., Brankovic-Sreckovic, V., et al., 2009. Japan Association of Childhood Leukemia Study. Pediatr. Int. 44,
Genetic risk factors for arterial ischemic stroke in children: a pos- 293–299.
1006.e2 PART III  Neurological Diseases and Their Treatment

Ijland, M.M., Pereira, R.R., Cornelissen, E.A., 2008. Incidence of late Mayer, T.O., Biller, J., O’Donnell, J., et al., 2002. Contrasting the neu-
vitamin K deficiency bleeding in newborns in the Netherlands in rologic complications of cardiac transplantation in adults and chil-
2005: evaluation of the current guideline. Eur. J. Pediatr. 167 (2), dren. J. Child Neurol. 17, 195–199.
165–169. Menkes, J., 2000. Metabolic diseases of the nervous system. In:
Janjua, N., Nasar, A., Lynch, J.K., et al., 2007. Thrombolysis for Menkes, J.H., Sarnat, H.B. (Eds.), Child Neurology, sixth ed. Lip-
ischemic stroke in children: data from the nationwide inpatient pincott Williams and Wilkins, Philadelphia, pp. 33–169.
sample. Stroke 38 (6), 1850–1854. Menkes, J., Fink, B., Hurvitz, C.C.H., et al., 2000. Neurologic manifes-
Jeanne, M., Labelle-Dumais, C., Jorgensen, J., et al., 2012. COL4A2 tations of systemic disease. In: Menkes, J.H., Sarnat, H.B. (Eds.),
mutations impair COL4A1 and COL4A2 secretion and cause hem- Child Neurology, sixth ed. Lippincott Williams and Wilkins, Phila-
orrhagic stroke. Am. J. Hum. Genet. 90 (1), 91–101. delphia, pp. 1093–1154.
Johnson, C.F., 2000. Abuse and neglect of children. In: Behrman, R.E., Moharir, M.D., Shroff, M., Stephens, D., et al., 2009. Anticoagulants
Kliegman, R.M., Jenson, H.B. (Eds.), Nelson Textbook of Pediatrics, in pediatric cerebral sinovenous thrombosis: a safety and outcome
sixteenth ed. Saunders, Philadelphia, p. 2414. study. Ann. Neurol. 67 (5), 590–599.
Jordan, L.C., Johnston, S.C., Wu, Y.W., et al., 2009. The importance of Mondal, D., Pradhan, L., Ali, M., et al., 2004. HAART drugs induce
cerebral aneurysms in childhood hemorrhagic stroke: a population- oxidative stress in human endothelial cells and increase endothelial
based study. Stroke 40 (2), 400–405. recruitment of mononuclear cells: exacerbation by inflammatory
Kenet, G., Lutkhoff, L.K., Albisetti, M., et al., 2010. Impact of cytokines and amelioration by antioxidants. Cardiovasc. Toxicol. 4,
thrombophilia on risk of arterial ischemic stroke or cerebral sino- 287–302.
venous thrombosis in neonates and children: a systematic review and Mueller, S., Sear, K., Hills, N.K., et al., 2013. Risk of first and recurrent
meta-analysis of observational studies. Circulation 121 (16), stroke in childhood cancer survivors treated with cranial and cervi-
1838–1847. cal radiation therapy. Int. J. Radiat. Oncol. Biol. Phys. 86 (4),
Khayat, A.S., Antunes, L.M., Guimaraes, A.C., et al., 2006. Cytotoxic 643–648.
and genotoxic monitoring of sickle cell anaemia patients treated Neuhaus, T.J., Goetschel, P., Schmugge, M., et al., 2000. Heparin-
with hydroxyurea. Clin. Exp. Med. 6, 33–37. induced thrombocytopenia type II on hemodialysis: switch to dana-
Kim, C.T., Han, J., Kim, H., 2009. Pediatric stroke recovery: a descrip- paroid. Pediatr. Nephrol. 14, 713–716.
tive analysis. Arch. Phys. Med. Rehabil. 90 (4), 657–662. Noje, C., Cohen, K., Jordan, L.C., 2013. Hemorrhagic and
Kirton, A., Crone, M., Benseler, S., et al., 2013. Fibromuscular dyspla- ischemic stroke in children with cancer. Pediatr. Neurol. 49 (4),
sia and childhood stroke. Brain 136, 1846–1856. 237–242.
Koch, H.G., Nabel, P., Junker, R., et al., 1999. The 677T genotype of the Normann, S., de Veber, G., Fobker, M., et al., 2009. Role of endog-
common MTHFR thermolabile variant and fasting homocysteine in enous testosterone concentration in pediatric stroke. Ann. Neurol.
childhood venous thrombosis. Eur. J. Pediatr. 158, S113–S116. 66 (6), 754–758.
Koga, Y., Akita, Y., Junko, N., et al., 2006. Endothelial dysfunction in Nowak-Göttl, U., Fiedler, B., Huge, A., et al., 2011. Plasma glutathione
MELAS improved by l-arginine supplementation. Neurology 66, peroxidase in pediatric stroke families. J. Thromb. Haemost. 9 (1),
1766–1769. 33–38.
Lagunju, I.A., Brown, B.J., Sodeinde, O.O., 2013. Stroke recurrence in Nowak-Göttl, U., Frohlich, B., Thedieck, S., et al., 2009. Association
Nigerian children with sickle cell disease treated with hydroxyurea. of the protein Z ATG haplotype with symptomatic nonvascular
Niger. Postgrad. Med. J. 20 (3), 181–187. stroke or thromboembolism in white children: a family-based
Lanthier, S., Lortie, A., Michaud, J., et al., 2001. Isolated angiitis of the cohort study. Blood 113 (10), 2336–2341.
CNS in children. Neurology 56, 837–842. Nowak-Göttl, U., Strater, R., Heincecke, A., et al., 1999. Lipoprotein
Laugesaar, R., Kolk, A., Tomberg, T., et al., 2007. Acutely and retro- (a) and genetic polymorphisms of clotting factor V, prothrombin,
spectively diagnosed perinatal stroke: a population-based study. and methylenetetrahydrofolate reductase are risk factors of sponta-
Stroke 38 (8), 2234–2240. neous ischemic stroke in childhood. Blood 94, 3678–3682.
Lee, J., Croen, L.A., Lindan, C., et al., 2005. Predictors of outcome in Papavasiliou, A., Bazigou-Fotopoulou, H., Ikeda, H., 2007. Familial
perinatal arterial stroke: a population-based study. Ann. Neurol. 58, moyamoya disease in two European children. J. Child Neurol. 22
303–308. (12), 1371–1376.
Lehman, L.L., Fullerton, H.J., 2013. Changing ethnic disparity in Pegelow, C.H., 2001. Stroke in children with sickle cell anaemia: aeti-
ischemic stroke mortality in US children after the STOP trial. JAMA ology and treatment. Paediatr. Drugs 3, 421–432.
Pediatr. 167 (8), 754–758. Pegelow, C.H., Macklin, E.A., Moser, F.G., et al., 2002. Longitudinal
Longo, N., Schrijver, I., Vogel, H., et al., 2008. Progressive cerebral changes in brain magnetic resonance imaging findings in children
vascular degeneration with mitochondrial encephalopathy. Am. J. with sickle cell disease. Blood 99, 3014–3018.
Med. Genet. A 146 (3), 361–367. Petaja, J., Peltola, K., Sairanen, H., et al., 1996. Fibrinolysis, anti-
Lynch, J.K., Nelson, K.B., 2001. Epidemiology of perinatal stroke. Curr. thrombin III, and protein C in neonates during cardiac operations.
Opin. Pediatr. 13, 499–505. J. Thorac. Cardiovasc. Surg. 112, 665–671.
Mader, I., Schoning, M., Klose, U., et al., 2002. Neonatal cerebral Pezzini, A., Grassi, M., Lodigiani, C., et al., 2014. Predictors of long-
infarction diagnosed by diffusion-weighted MRI: pseudonormaliza- term recurrent vascular events after ischemic stroke at young age:
tion occurs early. Stroke 33, 1142–1145. the Italian Project on Stroke in Young Adults. Circulation 129 (16),
Maguire, J.L., deVeber, G., Parkin, P.C., 2007. Association between 1668–1676.
iron-deficiency anemia and stroke in young children. Pediatrics 120 Rafay, M.F., Armstrong, D., Deveber, G., et al., 2006. Craniocervical
(5), 1053–1057. arterial dissection in children: clinical and radiographic presenta-
Mallick, A.A., Ganesan, V., Kirkham, F.J., et al., 2014. Childhood arte- tion and outcome. J. Child Neurol. 21, 8–16.
rial ischaemic stroke incidence, presenting features, and risk factors: Ranze, O., Ranze, P., Magnani, H.N., Greinacher, A., 1999. Heparin-
a prospective population-based study. Lancet Neurol. 13 (1), induced thrombocytopenia in paediatric patients—a review of the
35–43. literature and a new case treated with danaparoid sodium. Eur. J.
Mallick, A.A., Ganesan, V., O’Callaghan, F.J., 2009. Mortality from Pediatr. 158, S130–S133.
childhood stroke in England and Wales, 1921-2000. Arch. Dis. Raphael, J.L., Shetty, P.B., Liu, H., et al., 2008. A critical assessment of
Child. 95 (1), 12–19. transcranial Doppler screening rates in a large pediatric sickle cell
Mar, S., Kelly, J.E., Isbell, S., et al., 2013. Prevalence of white matter center: opportunities to improve healthcare quality. Pediatr. Blood
lesions and stroke in children with migraine. Neurology 81 (16), Cancer 51 (5), 647–651.
1387–1391. Ries, M., Gupta, S., Moore, D.F., et al., 2005. Pediatric Fabry disease.
Massicotte, P., Adams, M., Marzinotto, V., et al., 1996. Low-molecular- Pediatrics 115, e344–e355.
weight heparin in pediatric patients with thrombotic disease: a dose Risch, L., Huber, A.R., Schmugge, M., 2006. Diagnosis and treatment
finding study. J. Pediatr. 128, 313–318. of heparin-induced thrombocytopenia in neonates and children.
Matschke, J., Voss, J., Obi, N., et al., 2009. Nonaccidental head injury Thromb. Res. 118 (1), 123–135.
is the most common cause of subdural bleeding in infants <1 year Roach, E.S., 2000. Stroke in children. Curr. Treat. Options Neurol. 2,
of age. Pediatrics 124 (6), 1587–1594. 295–304.
Stroke in Children 1006.e3

Roach, E.S., Golomb, M.R., Adams, R., et al., 2008. Management of Strater, R., Kurnik, K., Heller, C., et al., 2001. Aspirin versus low-dose
stroke in infants and children. A scientific statement from a special low-molecular-weight heparin: antithrombotic therapy in pediatric 68
writing group of the American Heart Association Stroke Council ischemic stroke patients: a prospective follow-up study. Stroke 32,
and the Council on Cardiovascular Disease in the Young. Stroke 39, 2554–2558.
2644–2691. Tan, M.A., Miller, E., Shroff, M.M., et al., 2013. Alleviation of neonatal
Roaten, J.B., Patrick, D.A., Nydam, T.L., et al., 2006. Nonaccidental sinovenous compression to enhance cerebral venous blood flow. J.
trauma is a major cause of morbidity and mortality among patients Child Neurol. 28 (5), 583–588.
at a regional level 1 pediatric trauma center. J. Pediatr. Surg. 41 (12), Telfer, P., Coen, P., Chakravorty, S., et al., 2007. Clinical outcomes in
2013–2015. children with sickle cell disease living in England: a neonatal cohort
Rockett, S.R., Bryant, J.C., Morrow, W.R., et al., 2008. Preliminary in East London. Haematologica 92 (7), 905–912.
single center North American experience with the Berlin Heart pedi- Terwindt, G., Kors, E., Haan, J., et al., 2002. Mutation analysis of the
atric EXCOR device. ASAIO J. 54 (5), 479–482. CACNA1A calcium channel subunit gene in 27 patients with spo-
Rodan, L., McCrindle, B.W., Manlhiot, C., et al., 2012. Stroke recur- radic hemiplegic migraine. Arch. Neurol. 59, 1016–1018.
rence in children with congenital heart disease. Ann. Neurol. 72 (1), Thuillier, L., Chadefaux-Vekemans, B., Bonnefont, J.P., et al.,
103–111. 1998. Does the polymorphism 677C-T of the 5,10-
Rooks, V.J., Eaton, J.P., Ruess, L., et al., 2008. Prevalence and evolution methylenetetrahydrofolate reductase gene contribute to
of intracranial hemorrhage in asymptomatic term infants. Am. J. homocysteine-related vascular disease? J. Inherit. Metab. Dis. 21,
Neuroradiol. 29 (6), 1082–1089. 812–822.
Salih, M.A., Abdel-Gader, A.G., Al-Jarallah, A.A., et al., 2006a. Hema- Tian, H.L., Geng, Z., Cui, Y.H., et al., 2008. Risk factors for posttrau-
tologic risk factors for stroke in Saudi children. Saudi Med. J. 27, matic cerebral infarction in patients with moderate or severe head
S21–S34. trauma. Neurosurg. Rev. 31 (4), 431–436, discussion 436–437.
Salih, M.A., Abdel-Gader, A.G., Al-Jarallah, A.A., et al., 2006b. Infec- van den Tweel, X.W., Nederveen, A.J., Majoie, C.B., et al., 2009. Cer-
tious and inflammatory disorders of the circulatory system as risk ebral blood flow measurement in children with sickle cell disease
factors for stroke in Saudi children. Saudi Med. J. 27, S41–S52. using continuous arterial spin labeling at 3.0-Tesla MRI. Stroke 40
Salpietro, V., Mankad, K., Polizzi, A., et al., 2014. Pediatric Hashimo- (3), 795–800.
to’s encephalopathy with peripheral nervous system involvement. Vermylen, C., 2003. Hematopoietic stem cell transplantation in sickle
Pediatr. Int. 56, 413–416. cell disease. Blood Rev. 17, 163–166.
Schiffmann, R., 2001. Natural history of Fabry disease in males: pre- Volpe, J.J., 2001. Neurology of the Newborn. Saunders,
liminary observations. J. Inherit. Metab. Dis. 24, 15–17, discussion Philadelphia.
pp. 11–12. Wayne, A.S., Schoenike, S.E., Pegelow, C.H., 2000. Financial analysis
Schiffmann, R., Ries, M., Timmons, M., et al., 2006. Long-term therapy of chronic transfusion for stroke prevention in sickle cell disease.
with agalsidase alfa for Fabry disease: safety and effects on renal Blood 96, 2369–2372.
function in a home infusion setting. Nephrol. Dial. Transplant. 21, Weinberg, D.G., Rahme, R.J., Aoun, S.G., et al., 2011. Moyamoya
345–354. disease: functional and neurocognitive outcomes in the pediatric
Schulte, R., Jordan, L.C., Morad, A., Naftel, R.P., Wellons, J.C. 3rd, and adult populations. Neurosurg. Focus 30 (6), E21.
Sidonio, R., 2014. Rise in late onset vitamin K deficiency bleeding Wolff, V., Lauer, V., Rouyer, O., et al., 2011. Cannabis use, ischemic
in young infants because of omission or refusal of prophylaxis at stroke, and multifocal intracranial vasoconstriction: a prospective
birth. Pediatr. Neurol. 50, 564–568. study in 48 consecutive young patients. Stroke 42 (6),
Shah, S., Kumar, Y., McLean, B., et al., 2010. A dominantly inherited 1778–1780.
mutation in collagen IV A1 (COL4A1) causing childhood onset Zanon, E., Iorio, A., Rocino, A., et al., 2012. Intracranial haemorrhage
stroke without porencephaly. Eur. J. Paediatr. Neurol. 14 (2), in the Italian population of haemophilia patients with and without
182–187. inhibitors. 2012. Haemophilia 18 (1), 39–45.
Shürks, M., Rist, P.M., Bigal, M.E., et al., 2009. Migraine and cardio- Zhou, Q., Yang, D., Ombrello, A.K., et al., 2014. Early-onset stroke
vascular disease: systematic review and meta-analysis. BMJ 339, and vasculopathy associated with mutations in ADA2. N. Engl. J.
b3914. Med. 370 (10), 911–920.
Slooter, A.J., Rosendaal, F.R., Tanis, B.C., et al., 2005. Prothrombotic Zimmer, J.A., Garg, B.P., Williams, L.S., et al., 2007. Age-related varia-
conditions, oral contraceptives, and the risk of ischemic stroke. tion in presenting signs of childhood arterial ischemic stroke.
J. Thromb. Haemost. 3 (6), 1213–1217. Pediatr. Neurol. 37 (3), 171–175.
Starke, J.R., 1999. Tuberculosis of the central nervous system in chil- Zurin, A.A., Houkin, K., Asano, T., et al., 1997. Childhood ischemic
dren. Semin. Pediatr. Neurol. 6, 318–331. stroke caused by fibromuscular dysplasia of the intracranial artery—
Strater, R., Becker, S., von Eckhardstein, A., et al., 2002. Prospective case report. Neurol. Med. Chir. (Tokyo) 37 (7), 542–545.
assessment of risk factors for recurrent stroke during childhood—a
5-year follow-up study. Lancet 360, 1540–1545.

Вам также может понравиться