You are on page 1of 14

20 

Vascular Malformations (Arteriovenous


Malformations and Dural
Arteriovenous Fistulas)
SA MUEL KALB, BRADLEY A. GROSS, PETER NAKAJI

CLINICAL PEARLS
• Cerebral arteriovenous malformations (AVMs) are high-flow, connections between meningeal arteries and dural venous
high-pressure vascular anomalies that cause neurologic sinuses or leptomeningeal veins. Neurologic morbidity
morbidity, resulting in seizures, ischemic or steal symptoms, or associated with dAVFs can result in tinnitus, ocular symptoms,
hemorrhage. venous hypertension with neurologic deficits, and intracranial
• Cerebral AVMs are classified on the basis of treatment risk, hemorrhage.
including nidus size, location within eloquent or noneloquent • Cerebral dAVFs are classified by their risk of causing venous
tissue, and pattern of venous drainage (superficial versus hypertension or hemorrhage due to the presence or absence
deep). of cortical venous drainage, but they can also be influenced by
• Complete obliteration of AVMs is necessary to reduce the risk the presence of venous ectasia and the initial modality of
of bleeding and to mitigate the risk of seizures. The medical presentation.
literature lacks strong evidence that partial treatment confers • Spinal dAVFs are the most common type of spinal
protection from hemorrhage. Treatment modalities for cerebral arteriovenous shunt. They cause neurologic morbidity due to
or spinal AVMs include surgical resection, radiosurgery, venous hypertension. Surgical disconnection is associated with
endovascular occlusion, or a combination of these approaches. exceedingly high obliteration rates and low morbidity. Select
• Cerebral dural arteriovenous fistulas (dAVFs) are a distinct spinal dAVFs can be occluded endovascularly.
entity of vascular malformation composed of direct

Introduction with signs of spinal venous hypertension; these lesions are


composed of radicular artery feeders that shunt within the
Arteriovenous malformations (AVMs) and dural arteriovenous leaflets of the dura into an intradural vein at the junction of
fistulas (dAVFs) are the two most common types of arterio- the nerve root dura and the thecal sac.
venous shunts in the central nervous system. AVMs are a Since the 1960s, the quest to understand these lesions has
source of neurologic morbidity due to associated ischemic or led to the elucidation of many concepts in vascular physiology,
steal symptoms, hemorrhage, seizures (if cranial), or venous anatomy, and embryology. The management of both AVMs
hypertension (if spinal). AVMs are composed of pial or dural and dAVFs has also evolved considerably, with advancements
arterial vessels that tangle and coalesce into a nidus that shunts in neuroimaging, surgical techniques, and neuroendovascu-
blood flow into leptomeningeal veins in the absence of a capil- lar diagnostics and treatment procedures. Nevertheless, the
lary network. In contrast, patients with cerebral dAVFs may natural history and unpredictable course of both types of vas-
present with pulsatile tinnitus, ocular symptoms, or neurologic cular lesions continue to present a challenge for neurosurgeons
deficits secondary to venous hypertension. Cerebral dAVFs are and interventionalists, motivating multidisciplinary collab-
composed of meningeal arteries that shunt blood flow within orative approaches for a better understanding of the disease
the leaflets of the dura into a venous sinus or into the lepto- process in the hope of improving overall management and
meningeal veins. Patients with spinal dAVFs typically present outcomes.

313
Downloaded for John Egbuji (johnegbuji@gmail.com) at University of Otago from ClinicalKey.com.au by Elsevier on November 08, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
314 PART 3 Vascular Neurosurgery

Classification and Definitions


The formation of a vascular malformation within the central
nervous system is believed to be a dysplastic process. Four types
of dysplastic vascular malformations can be found in the
central nervous system: developmental venous anomalies, cap-
illary telangiectasias, cavernous malformations, and AVMs and
arteriovenous fistulas (AVFs). Developmental venous anoma-
lies are venous structures that drain normal brain tissue through
an abnormally straight venous channel that is usually deep and
fed by a cluster of veins typically referred to as a caput medusa
(Fig. 20.1A). Patients with developmental venous anomalies
are almost never symptomatic and should not be treated. Cap-
illary telangiectasias are small dysplastic capillary vessels that
are believed to be precursor lesions to cavernous malforma-
tions. Cavernous malformations are low-pressure, berry-like
vascular clusters of capillaries (Fig. 20.1B). They have a ten-
dency to bleed and to grow slowly. Over time, they often
provoke seizures or deficits from local tissue effects, which then
necessitates their surgical removal.
The last two categories of vascular malformations, AVMs
and dAVFs, are the main focus of this chapter. The character-
istic that separates AVMs and AVFs from other dysplastic
vascular malformations is the presence of an arteriovenous A
shunt through which oxygenated blood passes directly from
the arteries into the venous system without gas exchange in a
capillary bed. Unlike other vascular malformations, these
shunts are characterized by high blood flow and high-pressure
flow.
The three main types of AVMs found in the nervous system
are plexiform, fistulous, or a combination of the two. The
fistulous type (AVF) is present when an artery connects directly
to a vein without a capillary bed in between. AVFs most com-
monly occur within the dural leaflets in the central nervous
system as an acquired pathology termed a dAVF. Pial AVFs,
often considered congenital lesions, are far less common and
are typically discovered in younger patients. A plexiform mal-
formation (AVM) is an artery that connects to a network of
poorly differentiated, immature vessels (nidus) before reaching
a vein (Fig. 20.2).
There are numerous classification schemes for AVMs and
dAVFs. Although spinal arteriovenous shunt classifications B
have traditionally focused on the anatomy of the lesion itself,
• Figure 20.1  (A) On imaging, a developmental venous anomaly often
cerebral AVM classifications have historically aimed at helping
appears as a straight channel topped by a starburst of feeding veins. (B)
both the patient and the physician understand the risk of Illustration depicts the typical appearance of a cavernous malformation
various treatment modalities. Several classifications have been (arrow) as berry-like in configuration. Cavernous malformations can vary
proposed with the same goal: accuracy of treatment and ease considerably in size and location. (A, Used with permission from Barrow
of applicability.1,2 The Spetzler-Martin grading system3 (Fig. Neurological Institute, Phoenix, Arizona. B, Used with permission from
20.3) is the most widely used and the most commonly cited Thieme.)
classification for predicting the morbidity of surgical resection
of an AVM. It is based on three simple variables: size of the
malformation (1–3 points), the eloquence of the location (1 may be considered for conservative management because of the
point, if in an eloquent location), and the presence of deep high anticipated risk of surgical morbidity. It is important to
venous drainage (1 point, if present). The sum of all three emphasize that this classification scheme refers to surgical risk
variables provides the final grade, which ranges from I to V. and is based on the experience of a highly experienced neuro-
Patients with a grade I or II AVM tolerate resection with surgical team; external validity may thus be limited. In addi-
acceptable morbidity, whereas those with a grade IV or V AVM tion, other factors such as perforator supply and diffuse nidal

Downloaded for John Egbuji (johnegbuji@gmail.com) at University of Otago from ClinicalKey.com.au by Elsevier on November 08, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
CHAPTER 20  Vascular Malformations (Arteriovenous Malformations and Dural Arteriovenous Fistulas) 315

angioarchitecture are associated with increased surgical risk, equation: (0.1) × (volume in milliliters) + (0.02) × (age in
whereas younger patient age and AVM hemorrhage are associ- years) + 0.3 if the location is in the basal ganglia, thalamus, or
ated with lower surgical morbidity. These factors are not brainstem.5 AVM scores of less than 1 were associated with
considered in the original Spetzler-Martin grading scheme; obliteration and no radiosurgical morbidity in 89% of cases.
however, the Lawton supplementary scale incorporates some Similar to the Spetzler-Martin grading scale, the Virginia
of these factors.4 Radiosurgery AVM Scale assigns points for volume (0 points
Several grading schemes have been proposed to facilitate if less than 2 mL, 1 point if 2–4 mL, 2 points if more than
predicting the success of radiosurgical treatment of cerebral 4 mL) and eloquence (1 point). An additional point is added
AVMs; they account for AVM size and location. The for prior AVM hemorrhage. An AVM grade of 0 or 1 was
radiosurgery-based AVM scale also factors patient age into the associated with a morbidity-free AVM obliteration rate of 83%
and 79%, respectively.6
Cranial dAVF grading schemes help predict natural history
and are based on the original classification scheme by Djind-
jian.7 Type I dAVFs drain into a venous sinus and do not pose
a risk of venous hypertension or intracranial hemorrhage. Type
II dAVFs drain into a venous sinus with reflux into cortical
veins; thus they pose a risk of intracranial hemorrhage. Type
III dAVFs drain directly into cortical veins, and type IV dAVFs
drain into cortical veins with associated ectasia. Borden and
Shucart exclude the type IV dAVF and attempt to extrapolate
Djindjian’s scheme to spinal arteriovenous shunts; although
the Borden and Shucart scheme is not used for spinal lesions,
some use it instead of Djindjian’s today. Cognard and col-
leagues8 elaborated slightly on Djindjian’s scheme (Table 20.1).

Diagnostic Radiology
Noncontrast computed tomography (CT) of the head should
be the first imaging modality in any case of suspected intra-
• Figure 20.2  Illustration of a cerebral arteriovenous malformation, which
is characterized by feeding arteries (green arrows), a nidus, and draining cranial hemorrhage (Fig. 20.4). Although not useful for ruling
veins (blue arrow). (Used with permission from Barrow Neurological Insti- out the presence or absence of an AVM, noncontrast CT can
tute, Phoenix, Arizona.) demonstrate certain AVMs that produce calcium deposits,

TABLE
20.1  Cognard’s Modified Djindjian Scheme

Djindjian Borden-Shucart Cognard


Type I: Drainage into sinus or Type 1: Venous drainage directly into dural Type I: Venous drainage into dural venous sinus
meningeal vein venous sinus or meningeal vein with antegrade flow
Type II: Initial drainage into sinus with Type 2: Venous drainage into dural venous Type IIa: Venous drainage into dural venous
reflux into other sinuses or cortical sinus with CVR sinus with retrograde flow
veins
Type III: Initial drainage into cortical Type 3: Venous drainage directly into Type IIb: Venous drainage into dural venous
veins subarachnoid veins (CVR only) sinus with antegrade flow and CVR
Type IV: Initial drainage into cortical Type IIa+b: Venous drainage into dural venous
veins with giant venous pouch sinus with retrograde flow and CVR
Type III: Venous drainage directly into
subarachnoid veins (CVR only)
Type IV: Type III with venous ectasias of the
draining subarachnoid veins
Type V: Direct drainage into spinal perimedullary
veins

CVR, cortical venous reflux.

Downloaded for John Egbuji (johnegbuji@gmail.com) at University of Otago from ClinicalKey.com.au by Elsevier on November 08, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
316 PART 3 Vascular Neurosurgery

Spetzler-Ponce Grading System Spetzler-Martin Grading System

Grade
I
0 3 6
cm

Size - 1
Eloquence - 0
V. drainage - 0
Class
A

Grade
II
0 3 6 0 3 6 0 3 6
cm cm cm
Size - 1 Size - 1 Size - 2
Eloquence - 1 Eloquence - 0 Eloquence - 0
V. drainage - 0 V. drainage - 1 V. drainage - 0

Class Grade
B III
0 3 6 0 3 6 0 3 6
cm cm cm
Size - 1 Size - 2 Size - 2
Eloquence - 1 Eloquence - 1 Eloquence - 0
V. drainage - 1 V. drainage - 0 V. drainage - 1

Grade
IV
0 3 6 0 3 6 0 3 6
cm cm cm
Size - 2 Size - 3 Size - 3
Eloquence - 1 Eloquence - 1 Eloquence - 0
Class V. drainage - 1 V. drainage - 0 V. drainage - 1
C

Grade
V
0 3 6
cm
Size - 3
Eloquence - 1
V. drainage - 1 © 2017 Barrow

• Figure 20.3  The Spetzler-Martin and Spetzler-Ponce grading systems. The Spetzler-Martin system
divides arteriovenous malformations into five grades (see text). The Spetzler-Ponce system simplifies that
system into just three grades, lettered A to C. (Used with permission from Barrow Neurological Institute,
Phoenix, Arizona.)

which aids the diagnostic process. Contrast-enhanced CT can AVM nidus, identifies associated vessels, and aids in quantify-
demonstrate serpiginous vascular enhancement that is distinc- ing any embolic material present within the AVM. In addition,
tively typical of an AVM. In the case of a dAVF, abnormally CTA is useful for characterization and stereotactic localization
enlarged dural sinuses or draining cerebral veins can be seen of the AVM before surgical resection or radiosurgical treat-
after contrast administration. ment. In an AVF, the presence of asymmetrically visible and
CT angiography (CTA) is helpful for recognizing brain enlarged arterial feeding vessels has high sensitivity and speci-
AVMs (Fig. 20.5). CTA allows calculation of the volume of an ficity for the diagnosis of dAVFs. CTA can therefore be used

Downloaded for John Egbuji (johnegbuji@gmail.com) at University of Otago from ClinicalKey.com.au by Elsevier on November 08, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
CHAPTER 20  Vascular Malformations (Arteriovenous Malformations and Dural Arteriovenous Fistulas) 317

3 cm in size. The study emphasized the significantly greater


sensitivity of CTA in detecting AVM-associated aneurysms
compared to magnetic resonance imaging (MRI).9
MRI is a useful tool in the diagnosis and management of
AVMs (Fig. 20.6A). It can define complex structures such
as the AVM nidus while also localizing adjacent eloquent
structures, which is helpful for treatment planning. On
T2-weighted MRI, hypointense signals are indicative of flow
voids that represent the various feeding arteries, draining veins,
or vessels of the nidus itself. Peripheral to the nidus, hypoin-
tense signals on T2-weighted MRI and gradient echo MRI can
also show hemosiderin deposition, which indicates subclinical
hemorrhage. Magnetic resonance angiography and magnetic
resonance venography may be helpful in delineating the pres-
ence of flow in main vessels to and from the nidus. These
imaging techniques are noninvasive methods for determining
the progress of obliteration after radiosurgical or embolic
therapy. For dAVFs, MRI may show the presence of flow voids
on the surface of the brain or in the soft tissues. If the vascular
lesion is large enough, magnetic resonance angiography may
show an enlarged sinus and possible feeding and draining
• Figure 20.4  A plain axial computed tomogram showing hemorrhage. vessels. T2-weighted MRI is helpful in visualizing lesions with
Subtle evidence of calcification suggests an underlying arteriovenous associated venous ectasia.
malformation. (Used with permission from Barrow Neurological Institute,
Phoenix, Arizona.)
Angiography
Digital subtraction angiography is the gold standard imaging
technique for the diagnosis and angiographic evaluation of
cerebral AVMs and dAVFs (Fig. 20.6B–C). The following
factors should be identified, characterized, and evaluated on
these angiograms: arterial supply (with attention to the pres-
ence of perforator supply); nidus location, size, and architec-
ture (compact or diffuse); feeding artery and intranidal
aneurysms; and drainage pattern (deep or superficial; outflow
stenosis).
The arterial supply is visualized to define the anatomy of
the AVM and to potentially identify pedicles for embolization
if indicated. Early classification schemes focused on the number
of arterial suppliers to the AVM, although modern schemes
have not emphasized this angiographic factor.3,4 Periventricular
AVMs have a particular proclivity to recruit a choroidal arterial
supply, whereas surface AVMs may have a meningeal arterial
supply. A middle meningeal artery supply is a particularly
inviting pedicle for embolization when indicated. A careful
examination for the presence of a perforator supply is critical
for surgical planning. These pedicles can be a surgical nuisance;
coagulation and operative control of these vessels are critical
• Figure 20.5  A computed tomography angiogram of a large left-hemi- because the surgeon may have to continue following bleeding
sphere ruptured arteriovenous malformation (AVM) shows intraventricular vessels that retract into deep, often eloquent, tissue.10,11 Embo-
blood and the prominent tangle of vessels that constitute the AVM. (Used lization of these pedicles is a welcome adjunct, although embo-
with permission from Barrow Neurological Institute, Phoenix, Arizona.) lization can be challenging and limited by the small size and
tortuosity of the vessel.11
as a screening tool in patients with suspected fistulas. In one Defining the nidus location is important for predicting both
study of 125 cerebral AVMs, the sensitivity of CTA in detect- natural history and treatment risk for the AVM. Deep AVMs
ing AVMs was 90%, whereas that for magnetic resonance have a worse natural history than superficial lesions.12,13 Fur-
angiography was 74%. The sensitivity of CTA increased to thermore, the treatment of deep AVMs or AVMs in eloquent
96% for unruptured AVMs and to 100% for AVMs at least regions is associated with a greater risk and an often lower rate

Downloaded for John Egbuji (johnegbuji@gmail.com) at University of Otago from ClinicalKey.com.au by Elsevier on November 08, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
318 PART 3 Vascular Neurosurgery

A B

C
• Figure 20.6  (A) An axial T2-weighted magnetic resonance image (MRI) shows a left occipital arterio-
venous malformation (AVM). The AVM appears dark on T2-weighted MRI because of the presence of
“flow voids.” (B) A magnetic resonance angiogram of the same AVM shows only the vessels of the AVM,
with the brain not visible in the image. (C) An anteroposterior angiogram of the same AVM is shown for
comparison. (Used with permission from Barrow Neurological Institute, Phoenix, Arizona.)

of complete obliteration with any treatment modality.3,4,6,14,15 with greater risk and lower rates of complete obliteration for
Although modern studies have not shown a relationship any treatment modality.4,10
between nidus size and natural history risk,12,13 larger AVMs Feeding artery and intranidal aneurysms are important
are associated with greater risk and lower rates of complete angiographic components to identify for both natural history
obliteration for any treatment modality.3–6,14,16 Similarly, diffuse evaluation and treatment planning. These “high-risk” features
(as opposed to compact) nidus angioarchitecture is associated independently increase the prospective risk of intracranial

Downloaded for John Egbuji (johnegbuji@gmail.com) at University of Otago from ClinicalKey.com.au by Elsevier on November 08, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
CHAPTER 20  Vascular Malformations (Arteriovenous Malformations and Dural Arteriovenous Fistulas) 319

hemorrhage. Although they can be addressed during intra- rare and usually occurs only in patients with cervical dAVFs
operative resection, they can also be preoperatively embolized with intracranial drainage.
or treated with embolization alone in the case of inoperative
AVMs.15
The drainage pattern of AVMs is an important factor for Clinical Presentation of Arteriovenous
predicting both their natural history and their treatment risk. Malformations
Exclusive, deep venous drainage is an independent risk factor
for AVM hemorrhage.12,13 Although less substantiated by Hemorrhagic presentation of AVMs is linked to many ana-
natural history studies, venous outflow stenosis or obstruction tomic features. However, it is unknown whether those ana-
is also a risk factor. Deep venous drainage does not necessarily tomic features predict the likelihood of AVM rupture. Features
impact the risk of radiosurgery or embolization, but it has been that can increase the risk of hemorrhagic presentation are small
identified as a risk factor for surgical resection.3,4 lesion size, infratentorial location, a small number of draining
veins, and a high-pressure feeding artery.13,21–24 Associated
Epidemiology and Natural History aneurysms and hypertension also increase the risk of hemor-
rhagic presentation.13,25 The sequelae of AVM hemorrhage are
Across several studies, the prevalence of AVMs has ranged from dependent on the location of the lesion. A small, deep AVM
5 to 613 cases per 100,000.17,18 Multiple natural history studies may hemorrhage exclusively in the parenchyma, whereas a
consistently show the overall average patient age at AVM pre- cortical AVM may have a subarachnoid hemorrhage compo-
sentation to be in the third decade, without a significant sex nent. Intraventricular bleeding can occur from AVMs situated
predilection. Patients with AVMs present most commonly adjacent to the ventricles. Combinations of bleeding locations
after hemorrhage. One meta-analysis reported an overall hem- are also seen. Initial management of hemorrhagic presentations
orrhagic presentation rate of 52%12; patients presented with is similar to that of cerebral parenchymal hemorrhage. Patients
seizures in 27% of cases. Other potential presentation variables with dAVFs may present only with subarachnoid hemorrhage;
include headaches, ischemia, and steal symptoms, which can thus dAVFs should be considered in the differential diagnosis
also be an incidental finding. A meta-analysis reported an of nonaneurysmal bleeding. Patients with a subarachnoid hem-
overall prospective annual hemorrhage rate of 3%; for unrup- orrhage in the absence of intracranial aneurysms should
tured AVMs, the hemorrhage rate was 2.2%, and for ruptured undergo six-vessel cerebral angiography to rule out a dAVF.
AVMs, the rate was 4.5%, higher within the first year.12 Similar Seizures are the second most common presentation of
epidemiologic data were reported in a pooled analysis of 293 patients with AVMs, particularly when the AVM is in the
patients with spinal AVMs.12 Mean patient age was 29.1 years, supratentorial location. Approximately 15% to 30% of all
without a sex predilection. Half the patients presented with patients with AVMs present with a focal or generalized
hemorrhage, and the reported overall annual hemorrhage rate seizure.26–28 AVM features associated with epileptogenic pre-
was 4%. sentation include a cortical location of the nidus or feeding
A pooled analysis of 395 cerebral dAVFs demonstrated that, artery, location of the AVM in the middle cerebral artery
across a cohort of 141 low-risk fistulas (no cortical venous territory, the presence of varices in the venous drainage, and
drainage), mean patient age was 51 years, with a slight female association of a varix with absence of intranidal aneurysms.27,29
sex predilection (0.7 : 1, male : female).19 Rarely incidentally Other factors significantly associated with the onset of sei-
discovered, these fistulas often are found in patients who zures include AVMs fed by the external carotid artery and a
present with tinnitus or, if there is involvement of the cavern- temporal or parietal cortical location.29 AVM-related seizures
ous sinus, with ocular symptoms. They do not pose a risk of are thought to result from cortical irritation or remodeling
intracranial hemorrhage and rarely progress to cortical venous from ischemia, altered hemodynamics, mass effect, or micro-
drainage (1.4% of cases). Patients with high-risk dAVFs pre- hemorrhage. The epileptogenic presentation of AVMs has a
sented at a slightly older age (mean, 59 years) with a slight long-term risk profile for hemorrhage similar to that of non-
male sex predilection (1.5 : 1, male : female). In this cohort, epileptogenic types of AVM presentations. However, definitive
30% of patients presented with hemorrhage and 30% pre- surgical treatment may not be ideal because of a patient’s age
sented with signs of venous hypertension without hemorrhage. and comorbid conditions. Seizure control with antiepileptic
Other modalities of presentation include incidental, tinnitus, drugs is considered the first-line treatment and is used when
or ocular symptoms. The overall prospective annual hemor- surgical treatment is not feasible. A consultation with an epi-
rhage rate for this cohort was 6%. The rate increased to 21% leptologist for multidrug therapy is indicated in patients with
for type IV dAVFs (direct cortical venous drainage with venous medically refractory seizures. A neurologic consultation can
ectasia) and to 46% for lesions in patients who presented with confirm the location of the epileptic focus using clinical semi-
hemorrhage. The annual hemorrhage rate decreased to 2% for ology or tools such as electroencephalography, magnetoen-
high-risk fistulas that were found incidentally. cephalography, or single-photon emission CT. Because most
Patients with spinal dAVFs tend to present in their 60s; AVMs do not harbor functional tissue, the location of this
there is a known male sex predilection (2–3 males : 1 female).20 epileptic focus with respect to the actual AVM nidus may help
Symptoms at presentation are typically myelopathic in patients determine the best course of treatment, which may include
with spinal venous hypertension. Hemorrhage is exceedingly radiosurgery.30,31

Downloaded for John Egbuji (johnegbuji@gmail.com) at University of Otago from ClinicalKey.com.au by Elsevier on November 08, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
320 PART 3 Vascular Neurosurgery

Other clinical presentations of patients with AVMs are tion, progression, and recurrence after complete surgical resec-
headaches and focal neurologic deficits. Chronic headaches tion have been reported,10,35–37 negating the notion that AVMs
occur in 6% to 14% of patients who have no evidence of are static. They are indeed dynamic lesions capable of remodel-
rupture, especially in those with occipital AVMs, which may ing over time.
be a predisposing factor for rupture.26 The headaches can be
similar to migraines, localizing on one side of the brain, but Molecular Biology and Genetics
they may be more permanent in nature. However, this fact
does not necessarily preclude migraines as an independent Intracranial AVMs are associated with many factors known to
pathologic entity, and patients should be informed that the be present at higher concentrations than in normal tissue,34
goal of successful AVM treatment is the obliteration of some of which are observed in cerebral neoplastic processes.38,39
the nidus in order to mitigate the risk of a debilitating Further identification of associated angiogenic factors may
hemorrhage. allow for future targeted medical therapies. Around 900 genes
Neurologic deficits occur in 3% to 10% of patients who have been shown to have altered expression in AVMs.34,40
present with AVMs. Deficits may be transient, progressive, or However, a detailed discussion of the implicated molecular
permanent, and they can vary with the morphologic nature of factors is beyond the scope of this chapter. The factors with
the malformation. Deficits typically arise secondary to mass abnormally high levels of expression in AVMs are discussed
effect on adjacent structures or hemodynamic disturbances here.
such as the phenomenon of arterial steal. Arterial steal is the Vascular endothelial growth factor (VEGF) is present in
result of high-volume arteriovenous shunting that disrupts the increased amounts not only in AVMs but also in surrounding
vascular supply and regulation of normal brain structures by tissue. VEGF is normally expressed at high levels during
redirecting the blood flow toward the shunt at the expense of embryonic development, thus playing a key role in angiogen-
the normal vascular beds.31 The theory of arterial steal suggests esis, vascular proliferation, and capillary migration. VEGF
that large AVMs should correlate with increased physiologic expression is typically suppressed in adulthood but can be
evidence of steal. However, arterial steal is thought to be rela- rapidly increased by HIF-1 (hypoxia-induced factor) in
tively rare because the surrounding brain develops adaptive response to a low oxygen microenvironment, which stimulates
responses.31,32 the growth of AVMs.
Fibroblast growth factors are thought to assist with pro-
genitor cell differentiation to angioblasts during vasculogen-
Pathogenesis and Development of esis. The differentiation of fibroblasts to smooth muscle cells
Arteriovenous Malformations is likely regulated by basic fibroblast growth factor, which
is associated with the arterialization of AVM veins. Finally,
The pathogenesis of intracranial AVMs is not well elucidated. angiopoietins regulate the recruitment of smooth muscle
The traditional thought on AVM development leans toward a cells and the development of pericytes to endothelial cells,
congenital process, meaning that AVMs result from failures in both of which are thought to promote vascular stabiliza-
embryonic development and are present at birth. Embryonic tion during angiogenesis. Other angiogenic factors known to
vascular development is the successive combination of three contribute to AVM development are minor trauma, venous
processes: vasculogenesis, angiogenesis, and arteriogenesis.33,34 hypertension, exogenous growth factors, inflammation, and
Vasculogenesis refers to the differentiation of vascular progeni- infection.38,39,41,42
tor cells into angioblasts (the precursors to endothelium) and These factors suggest that AVMs develop in response to
the formation of a haphazard network of immature cells that an environmental insult during development or because of a
eventually fuse to form a primitive capillary plexus. Angiogen- genetic predisposition. Genetic alterations may be sporadic or
esis involves selective apoptosis and migration of supporting familial. Several genetic syndromes, such as Wyburn-Mason
vascular smooth muscle cells to form a stable vascular bed. The syndrome, Sturge-Weber disease, and ataxia-telangiectasia,
complex interactions between vasculogenesis and angiogenesis are associated with an increased predisposition to develop
in the embryonic stage require multiple steps for cell prolifera- AVMs.34
tion, migration, differentiation, and programmed destruction. Hereditary hemorrhagic telangiectasia, also known as
Any misstep during this stage could be the initiating factor in Osler-Weber-Rendu disease, is an autosomal dominant disor-
the formation of a congenital AVM. Arteriogenesis may con- der with variable penetrance. Genetic analysis has isolated two
tribute to observed growth and remodeling of AVMs from the mutations: HHT1 (chromosome 9q) and HHT2 (chromo-
fetal stage onward. Mediated by vascular wall shear stress, some 12q). The incidence of AVM with HHT1 mutations is
arteriogenesis is the likely mechanism by which an AVM with approximately 10 times that of HHT2, which demonstrates the
low resistance recruits additional blood supply over time. connection between genetic mutations and the development
The evidence supporting a congenital origin of AVMs is of AVMs.43 In addition to known hereditary disorders, AVMs
limited to the histologic resemblance between AVM structures often occur within families, suggesting a multifactorial genetic
and the developing embryonic vasculature and genetic links, patterning to their formation. Nonetheless, more than 95% of
such as those found in patients with hereditary hemorrhagic AVMs are sporadic, and HHT mutations account for only 3%
telangiectasia and Wyburg-Mason syndromes. De novo forma- of the total.44

Downloaded for John Egbuji (johnegbuji@gmail.com) at University of Otago from ClinicalKey.com.au by Elsevier on November 08, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
CHAPTER 20  Vascular Malformations (Arteriovenous Malformations and Dural Arteriovenous Fistulas) 321

Physiology therapies include decreasing the blood flow by obstructing the


feeding arteries, which is the therapeutic goal, and in contrast,
The hemodynamic properties of AVMs continue to be the decreasing the flow by obstructing veins, which increases
subject of observation and debate. AVM physiology includes the intranidal pressure, and therefore leads to a bleeding risk
that of the malformation itself and the surrounding brain (Fig. 20.7).
structures. The uncertainty surrounding AVM physiology The congenital nature of the AVM implies that sur-
stems from individual variability in AVM features. For this rounding brain tissue developed in the setting of low perfu-
chapter, AVM hemodynamics are discussed in terms of flow sion pressure; consequently, the vascular bed receives lower
rates and pressures through three structures—the feeding local arterial pressure. This observation is exacerbated by
artery, nidus, and draining vein—of an AVM. the reduction in vascular resistance caused by an enlarging
In this hypothetical model, any increase in blood flow to nidus and the resultant preferential blood flow toward the
one structure is the same in all structures. Flow increases with AVM. This dynamic can progress until the surrounding brain
higher feeding arterial pressure, decreasing draining pressure, is inadequately perfused, which causes neurologic dysfunc-
or decreasing vascular resistance upon nidus expansion. A tion. This phenomenon is known as vascular steal, but it is
nidus can expand as the diameter of existing vessels increases difficult to demonstrate definitively.31 Nonetheless, there is
or through angiogenesis, thereby reducing vascular resistance. some evidence to support neuropsychological dysfunction
As a result, feeding artery pressure decreases or its flow rate as the result of this phenomenon.45–47 Another consequence
increases until the establishment of a new equilibrium. For of low perfusion pressure in surrounding brain tissue is the
pressure to increase in a nidus, flow can be increased by increas- loss of vascular autoregulation in response to normal pres-
ing pressure in the feeding artery or flow can be decreased by sure changes. This loss becomes important after AVM resec-
increasing pressure (indirectly by increasing distal resistance) tion, when overall systemic vascular resistance is increased.
in the draining vein. This model does not consider many of The dysfunctional vasculature becomes hyperemic, with an
the complex properties of AVMs such as capacitance (increased increased risk for postresection hemorrhage. This phenome-
nidus flow with decreased resistance and pressure). It is impor- non is known as normal perfusion pressure breakthrough.48 The
tant to note that the hemodynamic consequences of AVMs are theory of normal perfusion pressure breakthrough is neces-
widely dependent on the cross-sectional area of the narrowest sary to consider in imaging modalities dependent on blood
communicating segment between arterial and venous circula- flow (eg, functional MRI), because it could produce false
tion. The primary hemodynamic considerations in AVM results.

A B
• Figure 20.7  (A) A lateral view left internal carotid artery angiogram shows the same occipital arterio-
venous malformation (AVM) as shown in Fig. 20.6. The angiogram shows normal vessels and the large
vessels of the AVM, which fill early compared to the regular circulation. (B) The arterial phase anteropos-
terior angiogram of the AVM shows it filling predominantly off middle cerebral artery branches with some
contribution from the anterior cerebral artery. (Used with permission from Barrow Neurological Institute,
Phoenix, Arizona.)

Downloaded for John Egbuji (johnegbuji@gmail.com) at University of Otago from ClinicalKey.com.au by Elsevier on November 08, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
322 PART 3 Vascular Neurosurgery

Pediatric Arteriovenous Malformations malformations range from progressive myelopathy (Foix-


Alajouanine syndrome) to acute excruciating back pain sec-
Spontaneous intracranial hemorrhage in pediatric patients is ondary to spinal subarachnoid hemorrhage (coup de poignard
strongly associated with AVMs.49,50 Nonetheless, children fare of Michon).
better than adults after microsurgical resection of AVMs, likely The original classification of spinal vascular malformation
because of the neural plasticity commonly observed in pediat- by Anson and Spetzler recognized four different angiographic
ric patients.51,52 Like adults with AVMs, children with AVMs types of malformations.62 Type I refers to dAVFs, the result
are most likely to present with hemorrhage or seizures.53 Inter- of a fistula between a radiculomeningeal artery and a radicu-
estingly, a newborn with an AVM may present with congestive lar vein. Type I is the most common type of malformation,
heart failure, which often happens when the malformation is making up 70% of all spinal vascular malformations, with a
in the vein of Galen. Congestive heart failure secondary to male predilection.68 The typical presentation depends on the
left-to-right cardiovascular shunting connotes a relatively poor location of the lesion, which is commonly found in the tho-
prognosis. Advances in pediatric critical care and endovascular racolumbar region,69 and it is the result of venous congestion
techniques are credited with improving long-term outcomes in and hypoperfusion. Patients with type I lesions rarely have
these patients.54 a hemorrhagic presentation. Type II lesions are intramedul-
The management of pediatric AVMs is similar to that of lary glomus AVMs with a vascular supply from the anterior
AVMs in adults, with a strong focus on aggressive surgical or posterior spinal circulation or both. Patients with these
resection of low-grade lesions to prevent future hemorrhage lesions, which are commonly found in the cervicomedullary
and recurrence. The residual diffuse-type AVM is prone to junction, typically present with acute neurolgic symptoms.
recur in pediatric patients.55,56 One possible explanation for Type III lesions are juvenile malformations that can be intra-
this phenomenon involves microshunting not apparent on medullary or extramedullary in location. Type IV AVMs are
angiography that enlarges over time. This idea is also consistent intradural, extramedullary lesions on the surface of the spinal
with the degree of arteriovenous shunting and the observation cord. Type IV lesions are the result of a fistula between a spinal
that many pediatric AVMs grow in volume over time. Most artery and spinal vein. An alternative method classifies spinal
pediatric patients are successfully treated with microsurgical vascular malformations into intramedullary or extramedullary
resection,53,56–58 which is associated with high rates of AVM locations.70
obliteration, as confirmed by angiography, and the low rate of
neurologic complications.58 Other therapies include radiosur- Treatment
gery and endovascular embolization. Endovascular interven-
tions are typically reserved for preoperative adjunct therapy, Surgeons use embolization of cerebral AVMs primarily as an
whereas radiosurgery is used for inoperable lesions.56 Stereo- adjunct to surgery, targeting high-risk features such as feeding
tactic radiosurgery is safe and effective in inoperable cases and artery or intranidal aneurysms (Fig. 20.8). Small, well-selected,
is most efficacious for small-volume AVMs located in critical inoperable, ruptured AVMs may be embolized with the
brain regions.59,60 Aggressive multimodality treatment (a goal of angiographic elimination of perceptible shunting.71,72
combination of microsurgery, radiosurgery, and endovascular N-butylcyanoacrylate (NBCA) and Onyx (ethylene-vinyl
embolization) for high-grade AVMs (Spetzler-Martin grade alcohol copolymer) are two typical embolizing agents that are
4–5) results in low cure rates and high complication rates.57 used and have similar associated morbidity.73 NBCA is a liquid
Thus prehemorrhagic high-grade lesions are best treated con- adhesive that rapidly polymerizes. It is ideal for high-flow
servatively and observed for progression.53 Predictors of poor shunts and for when a swift injection is needed.6,74 Onyx is
outcomes in pediatric patients include poor pretreatment func- a copolymer of ethylene-vinyl alcohol dissolved in dimethyl
tional status and high-grade and left-sided AVMs.57 sulfoxide that precipitates to form a nonadhesive solid.75
Detachable-tip microcatheters are now commonly used; they
Spinal Vascular Malformations may be intentionally glued in and then used for added nidal
penetration with Onyx via the pressure-cooker technique.76
Spinal vascular malformations are rare, making up 3% to 4% Onyx is used more often than NBCA for AVM embolization
of all space-occupying lesions in the spine.61 The classification because it allows for longer injections and superior penetra-
of spinal vascular malformations has been the subject of many tion.75,77 In our institutional experience with 342 AVMs embo-
debates and proposed classification systems.61–67 Spinal vascular lized over 446 sessions, most AVMs were embolized adjunctively
malformations are a heterogeneous group consisting of AVMs, before surgery. Procedural neurologic morbidity and mortality
dAVFs, hemangiomas, cavernous angiomas, and aneurysms. was 9.6% and 0.3%, respectively.77 Similar to the use of embo-
For simplicity, spinal vascular malformations can be divided lization in cerebral AVMs, the role of embolization for spinal
into extradural AVFs and intradural AVMs and AVFs. Extra- AVMs is mainly adjunctive for partial or full targeting of high-
dural AVFs are viewed as acquired, slow-flow lesions that risk features.12
develop over a long time. Extradural AVFs result in perimedul- In contrast to the use of embolization as an adjunct treat-
lary venous system dilation, stasis, and eventual hypoxia of the ment in cerebral AVMs, embolization is the initial treatment
spinal cord. In contrast, intradural AVFs and AVMs are con- of choice for cerebral dAVFs, with the intent of angiographic
genital lesions. The clinical presentations of spinal vascular obliteration. The introduction of Onyx was a landmark advance

Downloaded for John Egbuji (johnegbuji@gmail.com) at University of Otago from ClinicalKey.com.au by Elsevier on November 08, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
CHAPTER 20  Vascular Malformations (Arteriovenous Malformations and Dural Arteriovenous Fistulas) 323

A B

C D
• Figure 20.8  (A) A lateral internal carotid artery angiogram shows a temporal arteriovenous malforma-
tion (AVM) with a large nidal aneurysm, which was thought to be the source of the hemorrhage and hence
a good target for embolization. (B) The cast of the aneurysm left behind on the subtracted angiogram
(same view). (C) An angiogram obtained after the subtracted angiogram still shows the AVM, but the
aneurysm has been excluded from the circulation and is notably absent. (D) A plain computed tomogram
shows the hyperdense embolic material. (Used with permission from Barrow Neurological Institute,
Phoenix, Arizona.)

in the endovascular management of these lesions, allowing for the endovascular angiographic obliteration rate was 80% in
prolonged injections and better penetration of the draining the Onyx era. The rate of significant improvement or resolu-
vein, which is the goal of the procedure. Although indirect tion of tinnitus or ocular symptoms after initial treatment was
carotid-cavernous fistulas and selected marginal sinus fistu- also approximately 80%. The overall complication rate was
las are still often managed via transvenous coil embolization; 8%, with permanent neurologic morbidity occurring in 3% of
most other dAVFs are now typically managed via transarterial cases.78 Although surgery is an excellent and highly successful
Onyx embolization. In our published institutional experience modality for surgical disconnection of spinal dAVFs, emboliza-
with 260 dAVFs, excluding ethmoidal and torcular dAVFs, tion is, at times, an option, with feasible transarterial anatomy

Downloaded for John Egbuji (johnegbuji@gmail.com) at University of Otago from ClinicalKey.com.au by Elsevier on November 08, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
324 PART 3 Vascular Neurosurgery

for embolization seen in approximately 50% of patients.79 For neurologic deficits and is usually reserved for patients with
both procedures, the goal is disconnection of the AVF at its progressive symptoms or repeated hemorrhaging. Nonethe-
fistulous point. For the classic type I spinal dAVF feeding off less, some success with surgical resection has been docu-
a single pedicle, endovascular access and embolization of the mented. Compact “glomus” AVMs, in particular, can often
fistulous point at the nerve root sleeve can be curative. From a be circumferentially dissected and resected with good out-
surgical standpoint, cure is achieved by exposing and isolating comes. Other AVMs are more diffuse and may have func-
the lesion via a laminectomy. The surgeon then places a single tional spinal cord within them. Unlike cerebral AVMs in
clip on the appropriate feeding artery, which is usually suffi- which complete removal is an absolute condition of achieving
cient. Angiography and indocyanine green video angiography a surgical cure, some spinal AVMs can be “pruned” at the
are often used to confirm successful disconnection of the fistula. surface, leaving channels within the spinal cord parenchyma
The treatment of spinal AVMs is somewhat controversial. that no longer fill with blood and do not carry any risk of
Direct resection is considered to carry a very high risk of hemorrhage.

Summary
Vascular malformations in the brain have various pathophysiol- vary widely in their anatomy, size, and complexity. The choice
ogies, clinical presentations, prognostic implications, and treat- of treatment depends on the presentation, symptoms, and
ments. Some, such as developmental venous anomalies, should risks of treatment for the particular lesion. Neurosurgeons may
be recognized for their benign nature. AVFs are acquired lesions choose from a variety of treatment modalities including endo-
that are pathophysiologically simpler than AVMs and can be vascular treatment, radiosurgery, and microsurgery, and obser-
cured by disconnection. AVMs are congenital anomalies that vation alone may be an appropriate option for some patients.

Acknowledgments Gross BA, Du R. Natural history of cerebral arteriovenous malforma-


tions: a meta-analysis. J Neurosurg. 2013;118(2):437-443.
The authors thank the Neuroscience Publications staff of Lawton MT, Kim H, McCulloch CE, et al. A supplementary grading
Barrow Neurological Institute for help with manuscript prepa- scale for selecting patients with brain arteriovenous malformations for
ration and artwork and Soliman Oushy for his contributions surgery. Neurosurgery. 2010;66(4):702-713, discussion 713.
to the chapter. Loh Y, Duckwiler GR, Onyx Trial I. A prospective, multicenter, random-
ized trial of the Onyx liquid embolic system and N-butyl cyanoacry-
late embolization of cerebral arteriovenous malformations. Clinical
Selected Key References article. J Neurosurg. 2010;113(4):733-741.
Achrol AS, Guzman R, Varga M, et al. Pathogenesis and radiobiology of Mast H, Young WL, Koennecke HC, et al. Risk of spontaneous haemor-
brain arteriovenous malformations: implications for risk stratification rhage after diagnosis of cerebral arteriovenous malformation. Lancet.
in natural history and posttreatment course. Neurosurg Focus. 1997;350(9084):1065-1068.
2009;26(5):E9. Spetzler RF, Detwiler PW, Riina HA, et al. Modified classification of
Cognard C, Gobin YP, Pierot L, et al. Cerebral dural arteriovenous fis- spinal cord vascular lesions. J Neurosurg. 2002;96(2 suppl):145-156.
tulas: clinical and angiographic correlation with a revised classification Spetzler RF, Martin NA. A proposed grading system for arteriovenous
of venous drainage. Radiology. 1995;194(3):671-680. malformations. J Neurosurg. 1986;65(4):476-483.
Gross BA, Albuquerque FC, Moon K, et al. Evolution of treatment and Stefani MA, Porter PJ, terBrugge KG, et al. Large and deep brain arterio-
a detailed analysis of occlusion, recurrence, and clinical outcomes in venous malformations are associated with risk of future hemorrhage.
an endovascular library of 260 dural arteriovenous fistulas. J Neuro- Stroke. 2002;33(5):1220-1224.
surg. 2017;126(6):1884-1893. Please go to ExpertConsult.com to view the complete list of references.

Downloaded for John Egbuji (johnegbuji@gmail.com) at University of Otago from ClinicalKey.com.au by Elsevier on November 08, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
CHAPTER 20  Vascular Malformations (Arteriovenous Malformations and Dural Arteriovenous Fistulas) 324.e1

References 21. Kader A, Young WL, Pile-Spellman J, et al. The influence of


hemodynamic and anatomic factors on hemorrhage from cerebral
1. Luessenhop AJ, Gennarelli TA. Anatomical grading of supratentorial arteriovenous malformations. Neurosurgery. 1994;34(5):801-807,
arteriovenous malformations for determining operability. Neurosur- discussion 807-808.
gery. 1977;1(1):30-35. 22. Khaw AV, Mohr JP, Sciacca RR, et al. Association of infratentorial
2. Shi YQ, Chen XC. A proposed scheme for grading intracranial brain arteriovenous malformations with hemorrhage at initial pre-
arteriovenous malformations. J Neurosurg. 1986;65(4):484-489. sentation. Stroke. 2004;35(3):660-663.
3. Spetzler RF, Martin NA. A proposed grading system for arterio- 23. Langer DJ, Lasner TM, Hurst RW, et al. Hypertension, small size,
venous malformations. J Neurosurg. 1986;65(4):476-483. and deep venous drainage are associated with risk of hemorrhagic
4. Lawton MT, Kim H, McCulloch CE, et  al. A supplementary grading presentation of cerebral arteriovenous malformations. Neurosurgery.
scale for selecting patients with brain arteriovenous malformations 1998;42(3):481-486, discussion 487-489.
for surgery. Neurosurgery. 2010;66(4):702-713, discussion 713. 24. Stefani MA, Porter PJ, terBrugge KG, et al. Large and deep brain
5. Pollock BE, Flickinger JC. Modification of the radiosurgery- arteriovenous malformations are associated with risk of future hem-
based arteriovenous malformation grading system. Neurosurgery. orrhage. Stroke. 2002;33(5):1220-1224.
2008;63(2):239-243, discussion 243. 25. Langer RD, Pierce JJ, O’Hanlan KA, et al. Transvaginal ultraso-
6. Starke RM, Yen CP, Ding D, et al. A practical grading scale for pre- nography compared with endometrial biopsy for the detection of
dicting outcome after radiosurgery for arteriovenous malformations: endometrial disease: Postmenopausal Estrogen/Progestin Interven-
analysis of 1012 treated patients. J Neurosurg. 2013;119(4):981-987. tions Trial. N Engl J Med. 1997;337(25):1792-1798.
7. Djindjian R. Super-selective arteriography of branches of the exter- 26. Brown RD Jr, Wiebers DO, Torner JC, et al. Incidence and preva-
nal carotid artery. Surg Neurol. 1976;5(3):133-142. lence of intracranial vascular malformations in Olmsted County,
8. Cognard C, Gobin YP, Pierot L, et al. Cerebral dural arteriovenous Minnesota, 1965 to 1992. Neurology. 1996;46(4):949-952.
fistulas: clinical and angiographic correlation with a revised classifi- 27. Crawford PM, West CR, Chadwick DW, et al. Arteriovenous mal-
cation of venous drainage. Radiology. 1995;194(3):671-680. formations of the brain: natural history in unoperated patients. J
9. Gross BA, Frerichs KU, Du R. Sensitivity of CT angiography, Neurol Neurosurg Psychiatry. 1986;49(1):1-10.
T2-weighted MRI, and magnetic resonance angiography in detect- 28. Mast H, Young WL, Koennecke HC, et al. Risk of spontaneous
ing cerebral arteriovenous malformations and associated aneurysms. haemorrhage after diagnosis of cerebral arteriovenous malformation.
J Clin Neurosci. 2012;19(8):1093-1095. Lancet. 1997;350(9084):1065-1068.
10. Du R, Hashimoto T, Tihan T, et  al. Growth and regression of arterio- 29. Turjman F, Massoud TF, Sayre JW, et al. Epilepsy associated
venous malformations in a patient with hereditary hemorrhagic with cerebral arteriovenous malformations: a multivariate analy-
telangiectasia. Case report. J Neurosurg. 2007;106(3):470-477. sis of angioarchitectural characteristics. AJNR Am J Neuroradiol.
11. Gross BA, Duckworth EA, Getch CC, et al. Challenging traditional 1995;16(2):345-350.
beliefs: microsurgery for arteriovenous malformations of the basal 30. Lim YJ, Lee CY, Koh JS, et al. Seizure control of Gamma Knife
ganglia and thalamus. Neurosurgery. 2008;63(3):393-410, discussion radiosurgery for non-hemorrhagic arteriovenous malformations.
410-411. Acta Neurochir Suppl. 2006;99:97-101.
12. Gross BA, Du R. Spinal glomus (type II) arteriovenous malforma- 31. Mast H, Mohr JP, Osipov A, et al. “Steal” is an unestablished mecha-
tions: a pooled analysis of hemorrhage risk and results of interven- nism for the clinical presentation of cerebral arteriovenous malfor-
tion. Neurosurgery. 2013;72(1):25-32, discussion 32. mations. Stroke. 1995;26(7):1215-1220.
13. Stapf C, Mast H, Sciacca RR, et al. Predictors of hemorrhage in 32. Fink GR. Effects of cerebral angiomas on perifocal and remote
patients with untreated brain arteriovenous malformation. Neurol- tissue: a multivariate positron emission tomography study. Stroke.
ogy. 2006;66(9):1350-1355. 1992;23(8):1099-1105.
14. Dumont TM, Kan P, Snyder KV, et al. A proposed grading system 33. Heil M, Eitenmuller I, Schmitz-Rixen T, et al. Arteriogenesis
for endovascular treatment of cerebral arteriovenous malformations: versus angiogenesis: similarities and differences. J Cell Mol Med.
Buffalo score. Surg Neurol Int. 2015;6:3. 2006;10(1):45-55.
15. Pollock GA, Shaibani A, Awad I, et al. Intraventricular hemorrhage 34. Moftakhar P, Hauptman JS, Malkasian D, et al. Cerebral arterio-
secondary to intranidal aneurysm rupture-successful management venous malformations. Part 1: cellular and molecular biology. Neu-
by arteriovenous malformation embolization followed by intra- rosurg Focus. 2009;26(5):E10.
ventricular tissue plasminogen activator: case report. Neurosurgery. 35. Friedman JA, Pollock BE, Nichols DA. Development of a cerebral
2011;68(2):E581-E586, discussion E586. arteriovenous malformation documented in an adult by serial angi-
16. Heros RC, Korosue K, Diebold PM. Surgical excision of cerebral ography: case report. J Neurosurg. 2000;93(6):1058-1061.
arteriovenous malformations: late results. Neurosurgery. 1990;26(4): 36. Schmit BP, Burrows PE, Kuban K, et al. Acquired cerebral arterio-
570-577, discussion 577-578. venous malformation in a child with moyamoya disease: case report.
17. Laakso A, Dashti R, Seppanen J, et al. Long-term excess mortality in J Neurosurg. 1996;84(4):677-680.
623 patients with brain arteriovenous malformations. Neurosurgery. 37. Miyasaka Y, Nakahara K, Takagi H, et al. Development of mul-
2008;63(2):244-253, discussion 253-255. tiple cerebral arteriovenous malformations documented in an
18. Stapf C, Mohr JP, Pile-Spellman J, et al. Epidemiology and adult by serial angiography: case report. J Neurosurg. 2003;98(1):
natural history of arteriovenous malformations. Neurosurg Focus. 190-193.
2001;11(5):e1. 38. Chen Y, Pawlikowska L, Yao JS, et al. Interleukin-6 involvement
19. Gross BA, Du R. Natural history of cerebral arteriovenous malfor- in brain arteriovenous malformations. Ann Neurol. 2006;59(1):
mations: a meta-analysis. J Neurosurg. 2013;118(2):437-443. 72-80.
20. Muralidharan R, Saladino A, Lanzino G, et al. The clinical and 39. Chen Y, Zhu W, Bollen AW, et al. Evidence of inflammatory cell
radiological presentation of spinal dural arteriovenous fistula. Spine. involvement in brain arteriovenous malformations. Neurosurgery.
2011;36(25):E1641-E1647. 2008;62(6):1340-1349, discussion 1349-1350.

Downloaded for John Egbuji (johnegbuji@gmail.com) at University of Otago from ClinicalKey.com.au by Elsevier on November 08, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
324.e2 PART 3 Vascular Neurosurgery

40. Lim M, Cheshier S, Steinberg GK. New vessel formation in the 59. Dinca EB, de Lacy P, Yianni J, et al. Gamma knife surgery for pedi-
central nervous system during tumor growth, vascular malforma- atric arteriovenous malformations: a 25-year retrospective study. J
tions, and Moyamoya. Curr Neurovasc Res. 2006;3(3):237-245. Neurosurg Pediatr. 2012;10(5):445-450.
41. Hao Q, Zhu Y, Su H, et al. VEGF induces more severe cerebro- 60. Kano H, Kondziolka D, Flickinger JC, et al. Stereotactic radio-
vascular dysplasia in endoglin than in Alk1 mice. Transl Stroke Res. surgery for arteriovenous malformations, part 2: management of
2010;1(3):197-201. pediatric patients. J Neurosurg Pediatr. 2012;9(1):1-10.
42. Xu B, Wu YQ, Huey M, et al. Vascular endothelial growth factor 61. Bao YH, Ling F. Classification and therapeutic modalities of
induces abnormal microvasculature in the endoglin heterozygous spinal vascular malformations in 80 patients. Neurosurgery. 1997;
mouse brain. J Cereb Blood Flow Metab. 2004;24(2):237-244. 40(1):75-81.
43. Achrol AS, Guzman R, Varga M, et al. Pathogenesis and radiobi- 62. Anson JA, Spetzler RF. Interventional neuroradiology for spinal
ology of brain arteriovenous malformations: implications for risk pathology. Clin Neurosurg. 1992;39:388-417.
stratification in natural history and posttreatment course. Neurosurg 63. da Costa L, Dehdashti AR, terBrugge KG. Spinal cord vascular
Focus. 2009;26(5):E9. shunts: spinal cord vascular malformations and dural arteriovenous
44. Bharatha A, Faughnan ME, Kim H, et  al. Brain arteriovenous malfor- fistulas. Neurosurg Focus. 2009;26(1):E6.
mation multiplicity predicts the diagnosis of hereditary hemorrhagic 64. Grote EH, Voigt K. Clinical syndromes, natural history, and patho-
telangiectasia: quantitative assessment. Stroke. 2012;43(1):72-78. physiology of vascular lesions of the spinal cord. Neurosurg Clin N
45. Mahalick DM, Ruff RM, Hoi SU. Neuropsychological sequelae of Am. 1999;10(1):17-45.
arteriovenous malformations. Neurosurgery. 1991;29(3):351-357. 65. Heros RC, Debrun GM, Ojemann RG, et al. Direct spinal arterio-
46. Mahalick DM, Ruff RM, Heary RF, et al. Preoperative versus post- venous fistula: a new type of spinal AVM Case report. J Neurosurg.
operative neuropsychological sequelae of arteriovenous malforma- 1986;64(1):134-139.
tions. Neurosurgery. 1993;33(4):563-570, discussion 570-571. 66. Riche MC, Reizine D, Melki JP, et al. Classification of spinal cord
47. Marks MP, Lane B, Steinberg G, et al. Vascular characteristics of vascular malformations. Radiat Med. 1985;3(1):17-24.
intracerebral arteriovenous malformations in patients with clinical 67. Spetzler RF, Detwiler PW, Riina HA, et al. Modified classification of
steal. AJNR Am J Neuroradiol. 1991;12(3):489-496. spinal cord vascular lesions. J Neurosurg. 2002;96(2 suppl):145-156.
48. Spetzler RF, Wilson CB, Weinstein P, et al. Normal perfusion pres- 68. Patsalides A, Santillan A, Knopman J, et al. Endovascular man-
sure breakthrough theory. Clin Neurosurg. 1978;25:651-672. agement of spinal dural arteriovenous fistulas. J Neurointerv Surg.
49. Malik GM, Sadasivan B, Knighton RS, et al. The management 2011;3(1):80-84.
of arteriovenous malformations in children. Childs Nerv Syst. 69. Krings T. Vascular malformations of the spine and spinal cord*:
1991;7(1):43-47. anatomy, classification, treatment. Clin Neuroradiol. 2010;20(1):5-24.
50. Schoenberg BS, Mellinger JF, Schoenberg DG. Cerebrovascular 70. Minami S, Sagoh T, Nishimura K, et al. Spinal arteriovenous mal-
disease in infants and children: a study of incidence, clinical features, formation: MR imaging. Radiology. 1988;169(1):109-115.
and survival. Neurology. 1978;28(8):763-768. 71. Katsaridis V, Papagiannaki C, Aimar E. Curative embolization of
51. Fullerton HJ, Achrol AS, Johnston SC, et al. Long-term hemorrhage cerebral arteriovenous malformations (AVMs) with Onyx in 101
risk in children versus adults with brain arteriovenous malforma- patients. Neuroradiology. 2008;50(7):589-597.
tions. Stroke. 2005;36(10):2099-2104. 72. Mounayer C, Hammami N, Piotin M, et al. Nidal embolization of
52. Sanchez-Mejia RO, Chennupati SK, Gupta N, et al. Superior out- brain arteriovenous malformations using Onyx in 94 patients. AJNR
comes in children compared with adults after microsurgical resec- Am J Neuroradiol. 2007;28(3):518-523.
tion of brain arteriovenous malformations. J Neurosurg. 2006;105(2 73. Loh Y, Duckwiler GR, Onyx Trial I. A prospective, multicenter,
suppl):82-87. randomized trial of the Onyx liquid embolic system and N-butyl
53. Bristol RE, Albuquerque FC, Spetzler RF, et al. Surgical manage- cyanoacrylate embolization of cerebral arteriovenous malformations.
ment of arteriovenous malformations in children. J Neurosurg. Clinical article. J Neurosurg. 2010;113(4):733-741.
2006;105(2 suppl):88-93. 74. Haw CS, terBrugge K, Willinsky R, et al. Complications of embo-
54. Lasjaunias P, Hui F, Zerah M, et al. Cerebral arteriovenous mal- lization of arteriovenous malformations of the brain. J Neurosurg.
formations in children. Management of 179 consecutive cases and 2006;104(2):226-232.
review of the literature. Childs Nerv Syst. 1995;11(2):66-79, discus- 75. Weber W, Kis B, Siekmann R, et al. Preoperative embolization of
sion 79. intracranial arteriovenous malformations with Onyx. Neurosurgery.
55. Hladky JP, Lejeune JP, Blond S, et al. Cerebral arteriovenous 2007;61(2):244-252, discussion 252-254.
malformations in children: report on 62 cases. Childs Nerv Syst. 76. Chapot R, Stracke P, Velasco A, et al. The pressure cooker technique
1994;10(5):328-333. for the treatment of brain AVMs. J Neuroradiol. 2014;41(1):87-91.
56. Klimo P Jr, Rao G, Brockmeyer D. Pediatric arteriovenous mal- 77. Crowley RW, Ducruet AF, Kalani MY, et al. Neurological morbidity
formations: a 15-year experience with an emphasis on residual and and mortality associated with the endovascular treatment of cerebral
recurrent lesions. Childs Nerv Syst. 2007;23(1):31-37. arteriovenous malformations before and during the Onyx era. J
57. Darsaut TE, Guzman R, Marcellus ML, et al. Management of pedi- Neurosurg. 2015;122(6):1492-1497.
atric intracranial arteriovenous malformations: experience with mul- 78. Gross BA, Albuquerque FC, Moon K, et al. Evolution of treatment
timodality therapy. Neurosurgery. 2011;69(3):540-556, discussion and a detailed analysis of occlusion, recurrence, and clinical out-
556. comes in an endovascular library of 260 dural arteriovenous fistulas.
58. Gross BA, Storey A, Orbach DB, et al. Microsurgical treatment J Neurosurg. 2017;126(6):1884-1893.
of arteriovenous malformations in pediatric patients: the Boston 79. Gross BA, Albuquerque FC, Moon K, et al. Validation of an “endo-
Children’s Hospital experience. J Neurosurg Pediatr. 2015;15(1): vascular-first” approach to spinal dural arteriovenous fistulas: an
71-77. intention-to-treat analysis. J Neurointerv Surg. 2017;9(1):102-105.

Downloaded for John Egbuji (johnegbuji@gmail.com) at University of Otago from ClinicalKey.com.au by Elsevier on November 08, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.

You might also like